1
|
Bedoya Duque MA, Thomas WR, Dechmann DKN, Nieland J, Baldoni C, von Elverfeldt D, Muturi M, Corthals AP, Dávalos LM. Gene expression comparisons between captive and wild shrew brains reveal captivity effects. Biol Lett 2025; 21:20240478. [PMID: 39772919 PMCID: PMC11706642 DOI: 10.1098/rsbl.2024.0478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/17/2024] [Accepted: 11/15/2024] [Indexed: 01/11/2025] Open
Abstract
Compared with their free-ranging counterparts, wild animals in captivity experience different conditions with lasting physiological and behavioural effects. Although shifts in gene expression are expected to occur upstream of these phenotypes, we found no previous gene expression comparisons of captive versus free-ranging mammals. We assessed gene expression profiles of three brain regions (cortex, olfactory bulb and hippocampus) of wild shrews (Sorex araneus) compared with shrews kept in captivity for two months and undertook sample dropout to examine robustness given limited sample sizes. Consistent with captivity effects, we found hundreds of differentially expressed genes in all three brain regions, 104 overlapping across all three, that enriched pathways associated with neurodegenerative disease, oxidative phosphorylation and genes encoding ribosomal proteins. In the shrew, transcriptomic changes detected under captivity resemble responses in several human pathologies, including major depressive disorder and neurodegeneration. While interpretations of individual genes are tempered by small sample sizes, we propose captivity influences brain gene expression and function and can confound analyses of natural processes in wild individuals under captive conditions.
Collapse
Affiliation(s)
- Maria Alejandra Bedoya Duque
- Departamento de Ciencias Biológicas Bioprocesos y Biotecnología, Universidad Icesi, Cali, Valle del Cauca, Colombia
- Department of Ecology and Evolution, Stony Brook University, Stony Brook, NY, USA
| | - William R. Thomas
- Department of Ecology and Evolution, Stony Brook University, Stony Brook, NY, USA
| | - Dina K. N. Dechmann
- Max-Planck Institute for Animal Behavior, Radolfzell, Germany
- University of Konstanz, Konstanz, Germany
| | - John Nieland
- Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Cecilia Baldoni
- Max-Planck Institute for Animal Behavior, Radolfzell, Germany
- University of Konstanz, Konstanz, Germany
| | | | - Marion Muturi
- Max-Planck Institute for Animal Behavior, Radolfzell, Germany
- University of Konstanz, Konstanz, Germany
| | | | - Liliana M. Dávalos
- Department of Ecology and Evolution, Stony Brook University, Stony Brook, NY, USA
- Consortium for Inter-Disciplinary Environmental Research, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
2
|
Jayakanthan M, Manochkumar J, Efferth T, Ramamoorthy S. Lutein, a versatile carotenoid: Insight on neuroprotective potential and recent advances. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156185. [PMID: 39531935 DOI: 10.1016/j.phymed.2024.156185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/19/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Neurodegenerative diseases (NDDs) are a diverse group of neurological disorders with progressive neuronal loss at specific brain regions, leading to impaired cognitive functioning, loss of neuroplasticity, severe neurological impairment, and dementia. The incidence of neurodegenerative diseases is increasing at an alarming rate with current treatments struggling to barely prolong the inevitable. The desperation to discover a therapeutic agent to treat neurodegenerative diseases and to aid in the process of healthy recovery has opened a gateway into natural pigments. HYPOTHESIS The xanthophyll pigment lutein may bear the potential as a therapeutic agent against NDDs. RESULTS Lutein plays an important role in brain development, cognitive functioning, and improving neuroplasticity. In vitro and in vivo studies revealed the neuroprotective properties of lutein against NDDs such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and cerebral ischemia. The neuroprotective effect of lutein is evidenced by the reduction of free radicals and the simultaneous strengthening of the endogenous antioxidant systems by activating the NRF-2/ERK/AKT pathway. Further, it effectively suppressed mitochondrial aberrations, excitotoxicity, overaccumulation of metals, and its resultant complications. The immunomodulatory activity of lutein prevents neuroinflammation by hindering NF-κB nuclear translocation, regulation of NIK/IKK, PI3K/AKT, MAPK/ERK, JNK pathways, and ICAM-1 downregulation. Lutein also rescued the dysregulated cholinergic system and resolved memory defects. Along with its neuroprotective properties, lutein also improved neuroplasticity by enabling neurogenesis through increased GAP-43, NCAM, and BDNF levels. CONCLUSION Lutein exhibits strong neuroprotective activities against various NDDs. Though the investigations are in the exploratory phase, this review presents the consolidation of scattered evidence of the neuroprotective properties of lutein and urges its further exploration in clinical studies.
Collapse
Affiliation(s)
- Megha Jayakanthan
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Janani Manochkumar
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, 55128, Mainz, Germany
| | - Siva Ramamoorthy
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore 632014, India.
| |
Collapse
|
3
|
Bae HR, Shin SK, Lee JY, Choi SS, Kwon EY. Chronological Dynamics of Neuroinflammatory Responses in a High-Fat Diet Mouse Model. Int J Mol Sci 2024; 25:12834. [PMID: 39684545 DOI: 10.3390/ijms252312834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Obesity is known to affect various tissues and contribute to conditions such as neuroinflammation. However, the specific mechanisms and time-dependent progression of these effects across different tissues remain unclear. In this study, we monitored gene expression at intervals to examine the effects of a high-fat diet (HFD) on brain, liver, adipose, and muscle tissues in male C57/BJ mice, with a particular focus on neuroinflammation. Early inflammatory responses exhibit a progression that starts in the liver, extends to adipose tissue, and subsequently involves muscle and brain tissues. Although the brain did not show significant gene expression of inflammatory responses, mechanisms leading to neuroinflammation increased after 24 weeks, possibly through systemic chronic inflammation (SCI). Notably, mitochondrial complex I activity serves as a biomarker to indicate the inflammatory transition from the liver to adipose and other tissues caused by SCI. These similar gene expression dynamics were also observed in the hippocampus of Alzheimer's patients and in an Alzheimer's mouse model treated with a HFD. These results suggest that initially, the brain suppresses inflammatory responses, including interferon-gamma (IFN-γ), more than other tissues in response to a HFD. However, at the onset of SCI, the brain eventually exhibits inflammatory dynamics similar to those of other tissues. This underscores the significance of our findings, indicating that the early kinetics of chronic IFN-γ response and mitochondrial complex I activity inhibition serve as crucial biomarkers, emerging early in various conditions, including obesity and aging.
Collapse
Affiliation(s)
- Heekyong R Bae
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Su-Kyung Shin
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Ji-Yoon Lee
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Seong-Su Choi
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Eun-Young Kwon
- Department of Food Science and Nutrition, Kyungpook National University, Daegu 41566, Republic of Korea
- Center for Food and Nutritional Genomics, Kyungpook National University, Daegu 41566, Republic of Korea
- Center for Beautiful Aging, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
4
|
Bulduk BK, Tortajada J, Torres-Egurrola L, Valiente-Pallejà A, Martínez-Leal R, Vilella E, Torrell H, Muntané G, Martorell L. High frequency of mitochondrial DNA rearrangements in the peripheral blood of adults with intellectual disability. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2024:e13197. [PMID: 39506491 DOI: 10.1111/jir.13197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Mitochondrial DNA (mtDNA) rearrangements are recognised factors in mitochondrial disorders and ageing, but their involvement in neurodevelopmental disorders, particularly intellectual disability (ID) and autism spectrum disorder (ASD), remains poorly understood. Previous studies have reported mitochondrial dysfunction in individuals with both ID and ASD. The aim of this study was to investigate the prevalence of large-scale mtDNA rearrangements in ID and ID with comorbid ASD (ID-ASD). METHOD We used mtDNA-targeted next-generation sequencing and the MitoSAlt high-throughput computational pipeline in peripheral blood samples from 76 patients with ID (mean age 52.5 years, 37% female), 59 patients with ID-ASD (mean age 41.3 years, 46% female) and 32 healthy controls (mean age 42.4 years, 47% female) from Catalonia. RESULTS The study revealed a high frequency of mtDNA rearrangements in patients with ID, with 10/76 (13.2%) affected individuals. However, the prevalence was significantly lower in patients with ID-ASD 1/59 (1.7%) and in HC 1/32 (3.1%). Among the mtDNA rearrangements, six were identified as deletions (median size 6937 bp and median heteroplasmy level 2.3%) and six as duplications (median size 10 455 bp and median heteroplasmy level 1.9%). One of the duplications, MT-ATP6 m.8765-8793dup (29 bp), was present in four individuals with ID with a median heteroplasmy level of 3.9%. CONCLUSIONS Our results show that mtDNA rearrangements are frequent in patients with ID, but not in those with ID-ASD, when compared to HC. Additionally, MitoSAlt has demonstrated high sensitivity and accuracy in detecting mtDNA rearrangements, even at very low heteroplasmy levels in blood samples. While the high frequency of mtDNA rearrangements in ID is noteworthy, the role of these rearrangements is currently unclear and needs to be confirmed with further data, particularly in post-mitotic tissues and through age-matched control studies.
Collapse
Affiliation(s)
- B K Bulduk
- Àrea de Recerca, Hospital Universitari Institut Pere Mata (HUIPM), Reus, Catalonia, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV-CERCA), Universitat Rovira i Virgili (URV), Reus, Catalonia, Spain
| | - J Tortajada
- Àrea de Recerca, Hospital Universitari Institut Pere Mata (HUIPM), Reus, Catalonia, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV-CERCA), Universitat Rovira i Virgili (URV), Reus, Catalonia, Spain
| | - L Torres-Egurrola
- Àrea de Recerca, Hospital Universitari Institut Pere Mata (HUIPM), Reus, Catalonia, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV-CERCA), Universitat Rovira i Virgili (URV), Reus, Catalonia, Spain
| | - A Valiente-Pallejà
- Àrea de Recerca, Hospital Universitari Institut Pere Mata (HUIPM), Reus, Catalonia, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV-CERCA), Universitat Rovira i Virgili (URV), Reus, Catalonia, Spain
- CIBER de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - R Martínez-Leal
- Institut d'Investigació Sanitària Pere Virgili (IISPV-CERCA), Universitat Rovira i Virgili (URV), Reus, Catalonia, Spain
- CIBER de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
- Genètica i Ambient en Psiquiatria, Intellectual Disability and Developmental Disorders Research Unit (UNIVIDD), Fundació Villablanca, Reus, Catalonia, Spain
| | - E Vilella
- Àrea de Recerca, Hospital Universitari Institut Pere Mata (HUIPM), Reus, Catalonia, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV-CERCA), Universitat Rovira i Virgili (URV), Reus, Catalonia, Spain
- CIBER de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - H Torrell
- Centre for Omic Sciences (COS), Joint Unit Universitat Rovira i Virgili-EURECAT Technology Centre of Catalonia, Unique Scientific and Technical Infrastructures, Reus, Catalonia, Spain
| | - G Muntané
- Àrea de Recerca, Hospital Universitari Institut Pere Mata (HUIPM), Reus, Catalonia, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV-CERCA), Universitat Rovira i Virgili (URV), Reus, Catalonia, Spain
- CIBER de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
- Institut de Biologia Evolutiva (UPF-CSIC), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Parc de Recerca Biomèdica de Barcelona, Barcelona, Catalonia, Spain
| | - L Martorell
- Àrea de Recerca, Hospital Universitari Institut Pere Mata (HUIPM), Reus, Catalonia, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV-CERCA), Universitat Rovira i Virgili (URV), Reus, Catalonia, Spain
- CIBER de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Sonsungsan P, Aimauthon S, Sriwichai N, Namchaiw P. Unveiling mitochondria as central components driving cognitive decline in alzheimer's disease through cross-transcriptomic analysis of hippocampus and entorhinal cortex microarray datasets. Heliyon 2024; 10:e39378. [PMID: 39498000 PMCID: PMC11534180 DOI: 10.1016/j.heliyon.2024.e39378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/09/2024] [Accepted: 10/13/2024] [Indexed: 11/07/2024] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by symptoms such as memory loss and impaired learning. This study conducted a cross-transcriptomic analysis of AD using existing microarray datasets from the hippocampus (HC) and entorhinal cortex (EC), comparing them with age-matched non-AD controls. Both of these brain regions are critical for learning and memory processing and are vulnerable areas that exhibit abnormalities in early AD. The cross-transcriptomic analysis identified 564 significantly differentially expressed genes in HC and 479 in EC. Among these, 151 genes were significantly differentially expressed in both tissues, with functions related to synaptic vesicle clustering, synaptic vesicle exocytosis/endocytosis, mitochondrial ATP synthesis, hydrogen ion transmembrane transport, and structural constituent of cytoskeleton, suggesting a potential association between cognitive decline in AD, synaptic vesicle dynamics, dysregulation of cytoskeleton organization, and mitochondrial dysfunction. Further gene ontology analysis specific to the HC revealed the gene ontology enrichment in aerobic respiration, synaptic vesicle cycle, and oxidative phosphorylation. The enrichment analysis in CA1 of HC revealed differentiation in gene expression related to mitochondrial membrane functions involved in bioenergetics, mitochondrial electron transport, and biological processes associated with microtubule-based process, while analysis in the EC region showed enrichment in synaptic vesicle dynamics which is associated with neurotransmitter release and the regulation of postsynaptic membrane potential and synaptic transmission of GABAergic and glutamatergic synapse. Protein-protein interaction analysis highlighted central hub proteins predominantly expressed in mitochondria, involved in regulation of oxidative stress and ATP synthesis. These hub proteins interact not only within the mitochondria but also with proteins in the vesicular membrane and neuronal cytoskeleton, indicating a central role of mitochondria. This finding underscores the association between clinical symptoms and mitochondrial dysregulation of synaptic vesicle dynamics, cytoskeleton organization, and mitochondrial processes in both the HC and EC of AD. Therefore, targeting these dysregulated pathways could provide promising therapeutic targets aimed at cognitive decline and memory impairment in early AD stages.
Collapse
Affiliation(s)
- Pajaree Sonsungsan
- Mathematics and Statistics, School of Science, Walailak University, Nakhon Si Thammarat, Thailand
| | - Supatha Aimauthon
- Chemical Engineering, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
- Center for Biologics Research and Development, Chulabhorn Research Institute, Bangkok, Thailand
| | - Nattawet Sriwichai
- Center for Agricultural Systems Biology, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi (Bang Khun Thian), Bangkok, Thailand
| | - Poommaree Namchaiw
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
- Neuroscience Center for Research and Innovation, Learning Institute, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| |
Collapse
|
6
|
Venati SR, Uversky VN. Exploring Intrinsic Disorder in Human Synucleins and Associated Proteins. Int J Mol Sci 2024; 25:8399. [PMID: 39125972 PMCID: PMC11313516 DOI: 10.3390/ijms25158399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
In this work, we explored the intrinsic disorder status of the three members of the synuclein family of proteins-α-, β-, and γ-synucleins-and showed that although all three human synucleins are highly disordered, the highest levels of disorder are observed in γ-synuclein. Our analysis of the peculiarities of the amino acid sequences and modeled 3D structures of the human synuclein family members revealed that the pathological mutations A30P, E46K, H50Q, A53T, and A53E associated with the early onset of Parkinson's disease caused some increase in the local disorder propensity of human α-synuclein. A comparative sequence-based analysis of the synuclein proteins from various evolutionary distant species and evaluation of their levels of intrinsic disorder using a set of commonly used bioinformatics tools revealed that, irrespective of their origin, all members of the synuclein family analyzed in this study were predicted to be highly disordered proteins, indicating that their intrinsically disordered nature represents an evolutionary conserved and therefore functionally important feature. A detailed functional disorder analysis of the proteins in the interactomes of the human synuclein family members utilizing a set of commonly used disorder analysis tools showed that the human α-synuclein interactome has relatively higher levels of intrinsic disorder as compared with the interactomes of human β- and γ- synucleins and revealed that, relative to the β- and γ-synuclein interactomes, α-synuclein interactors are involved in a much broader spectrum of highly diversified functional pathways. Although proteins interacting with three human synucleins were characterized by highly diversified functionalities, this analysis also revealed that the interactors of three human synucleins were involved in three common functional pathways, such as the synaptic vesicle cycle, serotonergic synapse, and retrograde endocannabinoid signaling. Taken together, these observations highlight the critical importance of the intrinsic disorder of human synucleins and their interactors in various neuronal processes.
Collapse
Affiliation(s)
- Sriya Reddy Venati
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
- USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
7
|
Armirola-Ricaurte C, Zonnekein N, Koutsis G, Amor-Barris S, Pelayo-Negro AL, Atkinson D, Efthymiou S, Turchetti V, Dinopoulos A, Garcia A, Karakaya M, Moris G, Polat AI, Yiş U, Espinos C, Van de Vondel L, De Vriendt E, Karadima G, Wirth B, Hanna M, Houlden H, Berciano J, Jordanova A. Alternative splicing expands the clinical spectrum of NDUFS6-related mitochondrial disorders. Genet Med 2024; 26:101117. [PMID: 38459834 PMCID: PMC11180951 DOI: 10.1016/j.gim.2024.101117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024] Open
Abstract
PURPOSE We describe 3 families with Charcot-Marie-Tooth neuropathy (CMT), harboring a homozygous NDUFS6 NM_004553.6:c.309+5G>A variant previously linked to fatal Leigh syndrome. We aimed to characterize clinically and molecularly the newly identified patients and understand the mechanism underlying their milder phenotype. METHODS The patients underwent extensive clinical examinations. Exome sequencing was done in 4 affected individuals. The functional effect of the c.309+5G>A variant was investigated in patient-derived EBV-transformed lymphoblasts at the complementary DNA, protein, and mitochondrial level. Alternative splicing was evaluated using complementary DNA long-read sequencing. RESULTS All patients presented with early-onset, slowly progressive axonal CMT, and nystagmus; some exhibited additional central nervous system symptoms. The c.309+5G>A substitution caused the expression of aberrantly spliced transcripts and negligible levels of the canonical transcript. Immunoblotting showed reduced levels of mutant isoforms. No detectable defects in mitochondrial complex stability or bioenergetics were found. CONCLUSION We expand the clinical spectrum of NDUFS6-related mitochondrial disorders to include axonal CMT, emphasizing the clinical and pathophysiologic overlap between these 2 clinical entities. This work demonstrates the critical role that alternative splicing may play in modulating the severity of a genetic disorder, emphasizing the need for careful consideration when interpreting splice variants and their implications on disease prognosis.
Collapse
Affiliation(s)
- Camila Armirola-Ricaurte
- Molecular Neurogenomics group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium; Molecular Neurogenomics group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Noortje Zonnekein
- Molecular Neurogenomics group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium; Molecular Neurogenomics group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Georgios Koutsis
- Neurogenetics Unit, 1st Department of Neurology, Eginitio Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Silvia Amor-Barris
- Molecular Neurogenomics group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium; Molecular Neurogenomics group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Ana Lara Pelayo-Negro
- University Hospital Marqués de Valdecilla (IFIMAV), University of Cantabria, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Santander, Spain
| | - Derek Atkinson
- Molecular Neurogenomics group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium; Molecular Neurogenomics group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Stephanie Efthymiou
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Valentina Turchetti
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Argyris Dinopoulos
- 3rd Department of Pediatrics, Attiko Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonio Garcia
- Service of Clinical Neurophysiology, University Hospital Marqués de Valdecilla, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Santander, Spain
| | - Mert Karakaya
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Center for Rare Diseases, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - German Moris
- Service of Neurology, University Hospital Central de Asturias, University of Oviedo, Oviedo, Spain
| | - Ayşe Ipek Polat
- Department of Pediatric Neurology, Dokuz Eylül University, Izmir, Turkey
| | - Uluç Yiş
- Department of Pediatric Neurology, Dokuz Eylül University, Izmir, Turkey
| | - Carmen Espinos
- Rare Neurodegenerative Disease Laboratory, Centro de Investigación Príncipe Felipe (CIPF), CIBER on Rare Diseases (CIBERER), Valencia, Spain
| | - Liedewei Van de Vondel
- Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Laboratory of Neuromuscular Pathology, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Els De Vriendt
- Molecular Neurogenomics group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium; Molecular Neurogenomics group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Georgia Karadima
- Neurogenetics Unit, 1st Department of Neurology, Eginitio Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Brunhilde Wirth
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Center for Rare Diseases, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Michael Hanna
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Henry Houlden
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Jose Berciano
- University Hospital Marqués de Valdecilla (IFIMAV), University of Cantabria, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Santander, Spain
| | - Albena Jordanova
- Molecular Neurogenomics group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium; Molecular Neurogenomics group, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Department of Medical Chemistry and Biochemistry, Medical University-Sofia, Sofia, Bulgaria.
| |
Collapse
|
8
|
Key J, Gispert S, Auburger G. Knockout Mouse Studies Show That Mitochondrial CLPP Peptidase and CLPX Unfoldase Act in Matrix Condensates near IMM, as Fast Stress Response in Protein Assemblies for Transcript Processing, Translation, and Heme Production. Genes (Basel) 2024; 15:694. [PMID: 38927630 PMCID: PMC11202940 DOI: 10.3390/genes15060694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
LONP1 is the principal AAA+ unfoldase and bulk protease in the mitochondrial matrix, so its deletion causes embryonic lethality. The AAA+ unfoldase CLPX and the peptidase CLPP also act in the matrix, especially during stress periods, but their substrates are poorly defined. Mammalian CLPP deletion triggers infertility, deafness, growth retardation, and cGAS-STING-activated cytosolic innate immunity. CLPX mutations impair heme biosynthesis and heavy metal homeostasis. CLPP and CLPX are conserved from bacteria to humans, despite their secondary role in proteolysis. Based on recent proteomic-metabolomic evidence from knockout mice and patient cells, we propose that CLPP acts on phase-separated ribonucleoprotein granules and CLPX on multi-enzyme condensates as first-aid systems near the inner mitochondrial membrane. Trimming within assemblies, CLPP rescues stalled processes in mitoribosomes, mitochondrial RNA granules and nucleoids, and the D-foci-mediated degradation of toxic double-stranded mtRNA/mtDNA. Unfolding multi-enzyme condensates, CLPX maximizes PLP-dependent delta-transamination and rescues malformed nascent peptides. Overall, their actions occur in granules with multivalent or hydrophobic interactions, separated from the aqueous phase. Thus, the role of CLPXP in the matrix is compartment-selective, as other mitochondrial peptidases: MPPs at precursor import pores, m-AAA and i-AAA at either IMM face, PARL within the IMM, and OMA1/HTRA2 in the intermembrane space.
Collapse
Affiliation(s)
| | | | - Georg Auburger
- Experimental Neurology, Clinic of Neurology, University Hospital, Goethe University Frankfurt, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (J.K.); (S.G.)
| |
Collapse
|
9
|
Ahator SD, Hegstad K, Lentz CS, Johannessen M. Deciphering Staphylococcus aureus-host dynamics using dual activity-based protein profiling of ATP-interacting proteins. mSystems 2024; 9:e0017924. [PMID: 38656122 PMCID: PMC11097646 DOI: 10.1128/msystems.00179-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
The utilization of ATP within cells plays a fundamental role in cellular processes that are essential for the regulation of host-pathogen dynamics and the subsequent immune response. This study focuses on ATP-binding proteins to dissect the complex interplay between Staphylococcus aureus and human cells, particularly macrophages (THP-1) and keratinocytes (HaCaT), during an intracellular infection. A snapshot of the various protein activity and function is provided using a desthiobiotin-ATP probe, which targets ATP-interacting proteins. In S. aureus, we observe enrichment in pathways required for nutrient acquisition, biosynthesis and metabolism of amino acids, and energy metabolism when located inside human cells. Additionally, the direct profiling of the protein activity revealed specific adaptations of S. aureus to the keratinocytes and macrophages. Mapping the differentially activated proteins to biochemical pathways in the human cells with intracellular bacteria revealed cell-type-specific adaptations to bacterial challenges where THP-1 cells prioritized immune defenses, autophagic cell death, and inflammation. In contrast, HaCaT cells emphasized barrier integrity and immune activation. We also observe bacterial modulation of host processes and metabolic shifts. These findings offer valuable insights into the dynamics of S. aureus-host cell interactions, shedding light on modulating host immune responses to S. aureus, which could involve developing immunomodulatory therapies. IMPORTANCE This study uses a chemoproteomic approach to target active ATP-interacting proteins and examines the dynamic proteomic interactions between Staphylococcus aureus and human cell lines THP-1 and HaCaT. It uncovers the distinct responses of macrophages and keratinocytes during bacterial infection. S. aureus demonstrated a tailored response to the intracellular environment of each cell type and adaptation during exposure to professional and non-professional phagocytes. It also highlights strategies employed by S. aureus to persist within host cells. This study offers significant insights into the human cell response to S. aureus infection, illuminating the complex proteomic shifts that underlie the defense mechanisms of macrophages and keratinocytes. Notably, the study underscores the nuanced interplay between the host's metabolic reprogramming and immune strategy, suggesting potential therapeutic targets for enhancing host defense and inhibiting bacterial survival. The findings enhance our understanding of host-pathogen interactions and can inform the development of targeted therapies against S. aureus infections.
Collapse
Affiliation(s)
- Stephen Dela Ahator
- Centre for New Antibacterial Strategies (CANS) & Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Kristin Hegstad
- Centre for New Antibacterial Strategies (CANS) & Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT–The Arctic University of Norway, Tromsø, Norway
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
| | - Christian S. Lentz
- Centre for New Antibacterial Strategies (CANS) & Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT–The Arctic University of Norway, Tromsø, Norway
| | - Mona Johannessen
- Centre for New Antibacterial Strategies (CANS) & Research Group for Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT–The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
10
|
Liang KX, Chen A, Kianian A, Kristiansen CK, Yangzom T, Furriol J, Høyland LE, Ziegler M, Kråkenes T, Tzoulis C, Fang EF, Sullivan GJ, Bindoff LA. Activation of Neurotoxic Astrocytes Due to Mitochondrial Dysfunction Triggered by POLG Mutation. Int J Biol Sci 2024; 20:2860-2880. [PMID: 38904024 PMCID: PMC11186360 DOI: 10.7150/ijbs.93445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 05/03/2024] [Indexed: 06/22/2024] Open
Abstract
Mitochondrial diseases are associated with neuronal death and mtDNA depletion. Astrocytes respond to injury or stimuli and damage to the central nervous system. Neurodegeneration can cause astrocytes to activate and acquire toxic functions that induce neuronal death. However, astrocyte activation and its impact on neuronal homeostasis in mitochondrial disease remain to be explored. Using patient cells carrying POLG mutations, we generated iPSCs and then differentiated these into astrocytes. POLG astrocytes exhibited mitochondrial dysfunction including loss of mitochondrial membrane potential, energy failure, loss of complex I and IV, disturbed NAD+/NADH metabolism, and mtDNA depletion. Further, POLG derived astrocytes presented an A1-like reactive phenotype with increased proliferation, invasion, upregulation of pathways involved in response to stimulus, immune system process, cell proliferation and cell killing. Under direct and indirect co-culture with neurons, POLG astrocytes manifested a toxic effect leading to the death of neurons. We demonstrate that mitochondrial dysfunction caused by POLG mutations leads not only to intrinsic defects in energy metabolism affecting both neurons and astrocytes, but also to neurotoxic damage driven by astrocytes. These findings reveal a novel role for dysfunctional astrocytes that contribute to the pathogenesis of POLG diseases.
Collapse
Affiliation(s)
- Kristina Xiao Liang
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University Hospital, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
| | - Anbin Chen
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
- Department of Neurosurgery, Xinhua Hospital Affiliated toShanghai Jiaotong University School of Medicine, No. 1665, Kongjiang Road, 200092 Shanghai, China
| | - Atefeh Kianian
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
| | - Cecilie Katrin Kristiansen
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University Hospital, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
| | - Tsering Yangzom
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
| | - Jessica Furriol
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
| | - Lena Elise Høyland
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| | - Mathias Ziegler
- Department of Biomedicine, University of Bergen, Jonas Lies Vei 91, 5009 Bergen, Norway
| | - Torbjørn Kråkenes
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University Hospital, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
| | - Charalampos Tzoulis
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University Hospital, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
| | - Evandro Fei Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Oslo, Norway
- The Norwegian Centre on Healthy Ageing (NO-Age), 1478 Oslo, Norway
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, P. O. Box 1105, 0317 Oslo, Norway
| | - Gareth John Sullivan
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, P. O. Box 1105, 0317 Oslo, Norway
- Institute of Immunology, Oslo University Hospital, P.O. Box 4950, 0424 Oslo, Norway
- Hybrid Technology Hub Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, P. O. Box 1110, 0317 Oslo, Norway
- Department of Pediatric Research, Oslo University Hospital, P. O. Box 4950, 0424 Oslo, Norway
| | - Laurence A. Bindoff
- Department of Clinical Medicine (K1), University of Bergen, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
- Neuro-SysMed, Center of Excellence for Clinical Research in Neurological Diseases, Haukeland University Hospital, Jonas Lies vei 87, P. O. Box 7804, 5021 Bergen, Norway
| |
Collapse
|
11
|
Al Assi A, Posty S, Lamarche F, Chebel A, Guitton J, Cottet-Rousselle C, Prudent R, Lafanechère L, Giraud S, Dallemagne P, Suzanne P, Verney A, Genestier L, Castets M, Fontaine E, Billaud M, Cordier-Bussat M. A novel inhibitor of the mitochondrial respiratory complex I with uncoupling properties exerts potent antitumor activity. Cell Death Dis 2024; 15:311. [PMID: 38697987 PMCID: PMC11065874 DOI: 10.1038/s41419-024-06668-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 05/05/2024]
Abstract
Cancer cells are highly dependent on bioenergetic processes to support their growth and survival. Disruption of metabolic pathways, particularly by targeting the mitochondrial electron transport chain complexes (ETC-I to V) has become an attractive therapeutic strategy. As a result, the search for clinically effective new respiratory chain inhibitors with minimized adverse effects is a major goal. Here, we characterize a new OXPHOS inhibitor compound called MS-L6, which behaves as an inhibitor of ETC-I, combining inhibition of NADH oxidation and uncoupling effect. MS-L6 is effective on both intact and sub-mitochondrial particles, indicating that its efficacy does not depend on its accumulation within the mitochondria. MS-L6 reduces ATP synthesis and induces a metabolic shift with increased glucose consumption and lactate production in cancer cell lines. MS-L6 either dose-dependently inhibits cell proliferation or induces cell death in a variety of cancer cell lines, including B-cell and T-cell lymphomas as well as pediatric sarcoma. Ectopic expression of Saccharomyces cerevisiae NADH dehydrogenase (NDI-1) partially restores the viability of B-lymphoma cells treated with MS-L6, demonstrating that the inhibition of NADH oxidation is functionally linked to its cytotoxic effect. Furthermore, MS-L6 administration induces robust inhibition of lymphoma tumor growth in two murine xenograft models without toxicity. Thus, our data present MS-L6 as an inhibitor of OXPHOS, with a dual mechanism of action on the respiratory chain and with potent antitumor properties in preclinical models, positioning it as the pioneering member of a promising drug class to be evaluated for cancer therapy. MS-L6 exerts dual mitochondrial effects: ETC-I inhibition and uncoupling of OXPHOS. In cancer cells, MS-L6 inhibited ETC-I at least 5 times more than in isolated rat hepatocytes. These mitochondrial effects lead to energy collapse in cancer cells, resulting in proliferation arrest and cell death. In contrast, hepatocytes which completely and rapidly inactivated this molecule, restored their energy status and survived exposure to MS-L6 without apparent toxicity.
Collapse
Affiliation(s)
- Alaa Al Assi
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée (LBFA), Grenoble, France
| | - Solène Posty
- Cell death and Childhood Cancers Laboratory, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052- CNRS UMR5286, Université Claude Bernard de Lyon1, Centre Léon Bérard, LabEx DEVweCAN, Institut Convergence Plascan, Lyon, France
| | - Frédéric Lamarche
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée (LBFA), Grenoble, France
| | - Amel Chebel
- Centre International de Recherche en Infectiologie (Team LIB), Equipe labellisée La Ligue 2017 and 2023. Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Jérôme Guitton
- Laboratoire de biochimie et pharmacologie-toxicologie, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, F-69495, Pierre Bénite, France. Laboratoire de Toxicologie, Faculté de pharmacie ISPBL, Université Lyon 1, 69373, Lyon, France
| | - Cécile Cottet-Rousselle
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée (LBFA), Grenoble, France
| | - Renaud Prudent
- Université Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France
| | - Laurence Lafanechère
- Université Grenoble Alpes, Inserm U1209, CNRS UMR5309, Institute for Advanced Biosciences, Grenoble, France
| | - Stéphane Giraud
- Center for Drug Discovery and Development, Synergie Lyon Cancer Foundation, Lyon, Cancer Research Center, Centre Léon Bérard, Lyon, France
| | | | - Peggy Suzanne
- Normandie Univ., UNICAEN, CERMN, 14000, Caen, France
| | - Aurélie Verney
- Centre International de Recherche en Infectiologie (Team LIB), Equipe labellisée La Ligue 2017 and 2023. Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Laurent Genestier
- Centre International de Recherche en Infectiologie (Team LIB), Equipe labellisée La Ligue 2017 and 2023. Université Lyon, INSERM, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France
| | - Marie Castets
- Cell death and Childhood Cancers Laboratory, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052- CNRS UMR5286, Université Claude Bernard de Lyon1, Centre Léon Bérard, LabEx DEVweCAN, Institut Convergence Plascan, Lyon, France
| | - Eric Fontaine
- Université Grenoble Alpes, Inserm U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée (LBFA), Grenoble, France.
| | - Marc Billaud
- Cell death and Childhood Cancers Laboratory, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052- CNRS UMR5286, Université Claude Bernard de Lyon1, Centre Léon Bérard, LabEx DEVweCAN, Institut Convergence Plascan, Lyon, France.
| | - Martine Cordier-Bussat
- Cell death and Childhood Cancers Laboratory, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052- CNRS UMR5286, Université Claude Bernard de Lyon1, Centre Léon Bérard, LabEx DEVweCAN, Institut Convergence Plascan, Lyon, France.
| |
Collapse
|
12
|
Smith KK, Moreira JD, Wilson CR, Padera JO, Lamason AN, Xue L, Gopal DM, Flynn DB, Fetterman JL. A systematic review on the biochemical threshold of mitochondrial genetic variants. Genome Res 2024; 34:341-365. [PMID: 38627095 PMCID: PMC11067886 DOI: 10.1101/gr.278200.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 03/15/2024] [Indexed: 05/05/2024]
Abstract
Mitochondrial DNA (mtDNA) variants cause a range of diseases from severe pediatric syndromes to aging-related conditions. The percentage of mtDNA copies carrying a pathogenic variant, variant allele frequency (VAF), must reach a threshold before a biochemical defect occurs, termed the biochemical threshold. Whether the often-cited biochemical threshold of >60% VAF is similar across mtDNA variants and cell types is unclear. In our systematic review, we sought to identify the biochemical threshold of mtDNA variants in relation to VAF by human tissue/cell type. We used controlled vocabulary terms to identify articles measuring oxidative phosphorylation (OXPHOS) complex activities in relation to VAF. We identified 76 eligible publications, describing 69, 12, 16, and 49 cases for complexes I, III, IV, and V, respectively. Few studies evaluated OXPHOS activities in diverse tissue types, likely reflective of clinical access. A number of cases with similar VAFs for the same pathogenic variant had varying degrees of residual activity of the affected complex, alluding to the presence of modifying variants. Tissues and cells with VAFs <60% associated with low complex activities were described, suggesting the possibility of a biochemical threshold of <60%. Using Kendall rank correlation tests, the VAF of the m.8993T > G variant correlated with complex V activity in skeletal muscle (τ = -0.58, P = 0.01, n = 13); however, no correlation was observed in fibroblasts (P = 0.7, n = 9). Our systematic review highlights the need to investigate the biochemical threshold over a wider range of VAFs in disease-relevant cell types to better define the biochemical threshold for specific mtDNA variants.
Collapse
Affiliation(s)
- Karan K Smith
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Jesse D Moreira
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
- Programs in Human Physiology, Department of Health Sciences, Boston University Sargent College, Boston, Massachusetts 02215, USA
| | - Callum R Wilson
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - June O Padera
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Ashlee N Lamason
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Liying Xue
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Deepa M Gopal
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - David B Flynn
- Medical Sciences and Education, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA
| | - Jessica L Fetterman
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, USA;
| |
Collapse
|
13
|
Key J, Gispert S, Kandi AR, Heinz D, Hamann A, Osiewacz HD, Meierhofer D, Auburger G. CLPP-Null Eukaryotes with Excess Heme Biosynthesis Show Reduced L-arginine Levels, Probably via CLPX-Mediated OAT Activation. Biomolecules 2024; 14:241. [PMID: 38397478 PMCID: PMC10886707 DOI: 10.3390/biom14020241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/12/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
The serine peptidase CLPP is conserved among bacteria, chloroplasts, and mitochondria. In humans and mice, its loss causes Perrault syndrome, which presents with growth deficits, infertility, deafness, and ataxia. In the filamentous fungus Podospora anserina, CLPP loss leads to longevity. CLPP substrates are selected by CLPX, an AAA+ unfoldase. CLPX is known to target delta-aminolevulinic acid synthase (ALAS) to promote pyridoxal phosphate (PLP) binding. CLPX may also influence cofactor association with other enzymes. Here, the evaluation of P. anserina metabolomics highlighted a reduction in arginine/histidine levels. In Mus musculus cerebellum, reductions in arginine/histidine and citrulline occurred with a concomitant accumulation of the heme precursor protoporphyrin IX. This suggests that the increased biosynthesis of 5-carbon (C5) chain deltaALA consumes not only C4 succinyl-CoA and C1 glycine but also specific C5 delta amino acids. As enzymes responsible for these effects, the elevated abundance of CLPX and ALAS is paralleled by increased OAT (PLP-dependent, ornithine delta-aminotransferase) levels. Possibly as a consequence of altered C1 metabolism, the proteome profiles of P. anserina CLPP-null cells showed strong accumulation of a methyltransferase and two mitoribosomal large subunit factors. The reduced histidine levels may explain the previously observed metal interaction problems. As the main nitrogen-storing metabolite, a deficiency in arginine would affect the urea cycle and polyamine synthesis. Supplementation of arginine and histidine might rescue the growth deficits of CLPP-mutant patients.
Collapse
Affiliation(s)
- Jana Key
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (J.K.); (S.G.); (A.R.K.)
| | - Suzana Gispert
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (J.K.); (S.G.); (A.R.K.)
| | - Arvind Reddy Kandi
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (J.K.); (S.G.); (A.R.K.)
| | - Daniela Heinz
- Institute of Molecular Biosciences, Faculty of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt am Main, Germany; (D.H.); (A.H.); (H.D.O.)
| | - Andrea Hamann
- Institute of Molecular Biosciences, Faculty of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt am Main, Germany; (D.H.); (A.H.); (H.D.O.)
| | - Heinz D. Osiewacz
- Institute of Molecular Biosciences, Faculty of Biosciences, Goethe-University Frankfurt, 60438 Frankfurt am Main, Germany; (D.H.); (A.H.); (H.D.O.)
| | - David Meierhofer
- Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195 Berlin, Germany;
| | - Georg Auburger
- Goethe University Frankfurt, University Hospital, Clinic of Neurology, Experimental Neurology, Heinrich Hoffmann Str. 7, 60590 Frankfurt am Main, Germany; (J.K.); (S.G.); (A.R.K.)
| |
Collapse
|
14
|
Bennett NK, Lee M, Orr AL, Nakamura K. Systems-level analyses dissociate genetic regulators of reactive oxygen species and energy production. Proc Natl Acad Sci U S A 2024; 121:e2307904121. [PMID: 38207075 PMCID: PMC10801874 DOI: 10.1073/pnas.2307904121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 11/20/2023] [Indexed: 01/13/2024] Open
Abstract
Respiratory chain dysfunction can decrease ATP and increase reactive oxygen species (ROS) levels. Despite the importance of these metabolic parameters to a wide range of cellular functions and disease, we lack an integrated understanding of how they are differentially regulated. To address this question, we adapted a CRISPRi- and FACS-based platform to compare the effects of respiratory gene knockdown on ROS to their effects on ATP. Focusing on genes whose knockdown is known to decrease mitochondria-derived ATP, we showed that knockdown of genes in specific respiratory chain complexes (I, III, and CoQ10 biosynthesis) increased ROS, whereas knockdown of other low ATP hits either had no impact (mitochondrial ribosomal proteins) or actually decreased ROS (complex IV). Moreover, although shifting metabolic conditions profoundly altered mitochondria-derived ATP levels, it had little impact on mitochondrial or cytosolic ROS. In addition, knockdown of a subset of complex I subunits-including NDUFA8, NDUFB4, and NDUFS8-decreased complex I activity, mitochondria-derived ATP, and supercomplex level, but knockdown of these genes had differential effects on ROS. Conversely, we found an essential role for ether lipids in the dynamic regulation of mitochondrial ROS levels independent of ATP. Thus, our results identify specific metabolic regulators of cellular ATP and ROS balance that may help dissect the roles of these processes in disease and identify therapeutic strategies to independently target energy failure and oxidative stress.
Collapse
Affiliation(s)
- Neal K. Bennett
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
| | - Megan Lee
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Adam L. Orr
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY10021
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY10021
| | - Ken Nakamura
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA94158
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
- Graduate Program in Biomedical Sciences, University of California, San Francisco, CA94143
- Graduate Program in Neuroscience, University of California San Francisco, San Francisco, CA94158
- Department of Neurology, University of California, San Francisco, CA94158
| |
Collapse
|
15
|
Fergany A, Zong C, Ekuban FA, Wu B, Ueha S, Shichino S, Matsushima K, Iwakura Y, Ichihara S, Ichihara G. Transcriptome analysis of the cerebral cortex of acrylamide-exposed wild-type and IL-1β-knockout mice. Arch Toxicol 2024; 98:181-205. [PMID: 37971544 PMCID: PMC10761544 DOI: 10.1007/s00204-023-03627-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/12/2023] [Indexed: 11/19/2023]
Abstract
Acrylamide is an environmental electrophile that has been produced in large amounts for many years. There is concern about the adverse health effects of acrylamide exposure due to its widespread industrial use and also presence in commonly consumed foods and others. IL-1β is a key cytokine that protects the brain from inflammatory insults, but its role in acrylamide-induced neurotoxicity remains unknown. We reported recently that deletion of IL-1β gene exacerbates ACR-induced neurotoxicity in mice. The aim of this study was to identify genes or signaling pathway(s) involved in enhancement of ACR-induced neurotoxicity by IL-1β gene deletion or ACR-induced neurotoxicity to generate a hypothesis mechanism explaining ACR-induced neurotoxicity. C57BL/6 J wild-type and IL-1β KO mice were exposed to ACR at 0, 12.5, 25 mg/kg by oral gavage for 7 days/week for 4 weeks, followed by extraction of mRNA from mice cerebral cortex for RNA sequence analysis. IL-1β deletion altered the expression of genes involved in extracellular region, including upregulation of PFN1 gene related to amyotrophic lateral sclerosis and increased the expression of the opposite strand of IL-1β. Acrylamide exposure enhanced mitochondria oxidative phosphorylation, synapse and ribosome pathways, and activated various pathways of different neurodegenerative diseases, such as Alzheimer disease, Parkinson disease, Huntington disease, and prion disease. Protein network analysis suggested the involvement of different proteins in related to learning and cognitive function, such as Egr1, Egr2, Fos, Nr4a1, and Btg2. Our results identified possible pathways involved in IL-1β deletion-potentiated and ACR-induced neurotoxicity in mice.
Collapse
Affiliation(s)
- Alzahraa Fergany
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Building No. 15, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
- Laboratory of Genetics and Genetic Engineering in Department of Animal Husbandry and Animal Wealth Development, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Cai Zong
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Building No. 15, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Frederick Adams Ekuban
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Building No. 15, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Bin Wu
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Satoshi Ueha
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Kouji Matsushima
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Yoichiro Iwakura
- Division of Experimental Animal Immunology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Building No. 15, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan.
| |
Collapse
|
16
|
McGregor L, Acajjaoui S, Desfosses A, Saïdi M, Bacia-Verloop M, Schwarz JJ, Juyoux P, von Velsen J, Bowler MW, McCarthy AA, Kandiah E, Gutsche I, Soler-Lopez M. The assembly of the Mitochondrial Complex I Assembly complex uncovers a redox pathway coordination. Nat Commun 2023; 14:8248. [PMID: 38086790 PMCID: PMC10716376 DOI: 10.1038/s41467-023-43865-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
The Mitochondrial Complex I Assembly (MCIA) complex is essential for the biogenesis of respiratory Complex I (CI), the first enzyme in the respiratory chain, which has been linked to Alzheimer's disease (AD) pathogenesis. However, how MCIA facilitates CI assembly, and how it is linked with AD pathogenesis, is poorly understood. Here we report the structural basis of the complex formation between the MCIA subunits ECSIT and ACAD9. ECSIT binding induces a major conformational change in the FAD-binding loop of ACAD9, releasing the FAD cofactor and converting ACAD9 from a fatty acid β-oxidation (FAO) enzyme to a CI assembly factor. We provide evidence that ECSIT phosphorylation downregulates its association with ACAD9 and is reduced in neuronal cells upon exposure to amyloid-β (Aβ) oligomers. These findings advance our understanding of the MCIA complex assembly and suggest a possible role for ECSIT in the reprogramming of bioenergetic pathways linked to Aβ toxicity, a hallmark of AD.
Collapse
Affiliation(s)
- Lindsay McGregor
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 38043, Grenoble, France
| | - Samira Acajjaoui
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 38043, Grenoble, France
| | - Ambroise Desfosses
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS (IBS), 38044, Grenoble, France
| | - Melissa Saïdi
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 38043, Grenoble, France
| | - Maria Bacia-Verloop
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS (IBS), 38044, Grenoble, France
| | - Jennifer J Schwarz
- European Molecular Biology Laboratory (EMBL), 69117, Heidelberg, Germany
| | - Pauline Juyoux
- European Molecular Biology Laboratory (EMBL), 38043, Grenoble, France
| | - Jill von Velsen
- European Molecular Biology Laboratory (EMBL), 38043, Grenoble, France
| | - Matthew W Bowler
- European Molecular Biology Laboratory (EMBL), 38043, Grenoble, France
| | - Andrew A McCarthy
- European Molecular Biology Laboratory (EMBL), 38043, Grenoble, France
| | - Eaazhisai Kandiah
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 38043, Grenoble, France
| | - Irina Gutsche
- Institut de Biologie Structurale, Université Grenoble Alpes, CEA, CNRS (IBS), 38044, Grenoble, France.
- Department of Chemistry, Umeå University, Umeå, Sweden.
| | - Montserrat Soler-Lopez
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 38043, Grenoble, France.
| |
Collapse
|
17
|
Schmidt S, Stautner C, Vu DT, Heinz A, Regensburger M, Karayel O, Trümbach D, Artati A, Kaltenhäuser S, Nassef MZ, Hembach S, Steinert L, Winner B, Jürgen W, Jastroch M, Luecken MD, Theis FJ, Westmeyer GG, Adamski J, Mann M, Hiller K, Giesert F, Vogt Weisenhorn DM, Wurst W. A reversible state of hypometabolism in a human cellular model of sporadic Parkinson's disease. Nat Commun 2023; 14:7674. [PMID: 37996418 PMCID: PMC10667251 DOI: 10.1038/s41467-023-42862-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 10/23/2023] [Indexed: 11/25/2023] Open
Abstract
Sporadic Parkinson's Disease (sPD) is a progressive neurodegenerative disorder caused by multiple genetic and environmental factors. Mitochondrial dysfunction is one contributing factor, but its role at different stages of disease progression is not fully understood. Here, we showed that neural precursor cells and dopaminergic neurons derived from induced pluripotent stem cells (hiPSCs) from sPD patients exhibited a hypometabolism. Further analysis based on transcriptomics, proteomics, and metabolomics identified the citric acid cycle, specifically the α-ketoglutarate dehydrogenase complex (OGDHC), as bottleneck in sPD metabolism. A follow-up study of the patients approximately 10 years after initial biopsy demonstrated a correlation between OGDHC activity in our cellular model and the disease progression. In addition, the alterations in cellular metabolism observed in our cellular model were restored by interfering with the enhanced SHH signal transduction in sPD. Thus, inhibiting overactive SHH signaling may have potential as neuroprotective therapy during early stages of sPD.
Collapse
Affiliation(s)
- Sebastian Schmidt
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.
- Munich Institute of Biomedical Engineering, Department of Chemistry, Technical University of Munich, Munich, Germany.
| | - Constantin Stautner
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Duc Tung Vu
- Department for Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Alexander Heinz
- Department of Bioinformatics and Biochemistry and Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Martin Regensburger
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ozge Karayel
- Department for Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Dietrich Trümbach
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Anna Artati
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, Neuherberg, Germany
| | - Sabine Kaltenhäuser
- Department of Bioinformatics and Biochemistry and Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Mohamed Zakaria Nassef
- Department of Bioinformatics and Biochemistry and Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Sina Hembach
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Letyfee Steinert
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Beate Winner
- Department of Stem Cell Biology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Winkler Jürgen
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Martin Jastroch
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Malte D Luecken
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- Department of Mathematics, Technische Universität München, Garching bei München, Germany
| | - Gil Gregor Westmeyer
- Munich Institute of Biomedical Engineering, Department of Chemistry, Technical University of Munich, Munich, Germany
- Institute for Synthetic Biomedicine, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Matthias Mann
- Department for Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry and Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Florian Giesert
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | | | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany.
- Chair of Developmental Genetics, Munich School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE) site Munich, Munich, Germany.
| |
Collapse
|
18
|
Nicol T, Falcone S, Blease A, Vikhe P, Civiletto G, Omairi SS, Viscomi C, Patel K, Potter PK. Tissue-specific differences in the assembly of mitochondrial Complex I are revealed by a novel ENU mutation in ECSIT. Cardiovasc Res 2023; 119:2213-2229. [PMID: 37395010 PMCID: PMC10578914 DOI: 10.1093/cvr/cvad101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 03/26/2023] [Accepted: 04/19/2023] [Indexed: 07/04/2023] Open
Abstract
AIMS Mitochondrial Complex I assembly (MCIA) is a multi-step process that necessitates the involvement of a variety of assembly factors and chaperones to ensure that the final active enzyme is correctly assembled. The role of the assembly factor evolutionarily conserved signalling intermediate in the toll (ECSIT) pathway was studied across various murine tissues to determine its role in this process and how this varied between tissues of varying energetic demands. We hypothesized that many of the known functions of ECSIT were unhindered by the introduction of an ENU-induced mutation, while its role in Complex I assembly was affected on a tissue-specific basis. METHODS AND RESULTS Here, we describe a mutation in the MCIA factor ECSIT that reveals tissue-specific requirements for ECSIT in Complex I assembly. MCIA is a multi-step process dependent on assembly factors that organize and arrange the individual subunits, allowing for their incorporation into the complete enzyme complex. We have identified an ENU-induced mutation in ECSIT (N209I) that exhibits a profound effect on Complex I component expression and assembly in heart tissue, resulting in hypertrophic cardiomyopathy in the absence of other phenotypes. The dysfunction of Complex I appears to be cardiac specific, leading to a loss of mitochondrial output as measured by Seahorse extracellular flux and various biochemical assays in heart tissue, while mitochondria from other tissues were unaffected. CONCLUSIONS These data suggest that the mechanisms underlying Complex I assembly and activity may have tissue-specific elements tailored to the specific demands of cells and tissues. Our data suggest that tissues with high-energy demands, such as the heart, may utilize assembly factors in different ways to low-energy tissues in order to improve mitochondrial output. These data have implications for the diagnosis and treatment of various disorders of mitochondrial function as well as cardiac hypertrophy with no identifiable underlying genetic cause.
Collapse
Affiliation(s)
- Thomas Nicol
- Mammalian Genetics Unit, MRC Harwell Institute, Becquerel Avenue, Didcot, OX11 0RD, Oxfordshire, UK
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Sara Falcone
- Mammalian Genetics Unit, MRC Harwell Institute, Becquerel Avenue, Didcot, OX11 0RD, Oxfordshire, UK
- Centre for Cellular and Molecular Physiology, University of Oxford, Oxford, UK
| | - Andrew Blease
- Mammalian Genetics Unit, MRC Harwell Institute, Becquerel Avenue, Didcot, OX11 0RD, Oxfordshire, UK
| | - Pratik Vikhe
- Mammalian Genetics Unit, MRC Harwell Institute, Becquerel Avenue, Didcot, OX11 0RD, Oxfordshire, UK
| | | | | | - Carlo Viscomi
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK
| | - Paul K Potter
- Mammalian Genetics Unit, MRC Harwell Institute, Becquerel Avenue, Didcot, OX11 0RD, Oxfordshire, UK
- Department Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, UK
| |
Collapse
|
19
|
Serrano-Lorenzo P, Gobelli D, Garrido-Moraga R, Esteban-Amo MJ, López-López JR, Orduña A, de la Fuente MA, Martín MA, Simarro M. Development of a novel in vitro model to study the modulatory role of the respiratory complex I in macrophage effector functions. PLoS One 2023; 18:e0291442. [PMID: 37725617 PMCID: PMC10508620 DOI: 10.1371/journal.pone.0291442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023] Open
Abstract
Increasing evidence demonstrate that the electron transfer chain plays a critical role in controlling the effector functions of macrophages. In this work, we have generated a Ndufs4-/- murine macrophage cell lines. The Ndufs4 gene, which encodes a supernumerary subunit of complex I, is a mutational hotspot in Leigh syndrome patients. Ndufs4-/- macrophages showed decreased complex I activity, altered complex I assembly, and lower levels of maximal respiration and ATP production. These mitochondrial respiration alterations were associated with a shift towards a pro-inflammatory cytokine profile after lipopolysaccharide challenge and improved ability to phagocytose Gram-negative bacteria.
Collapse
Affiliation(s)
- Pablo Serrano-Lorenzo
- Hospital 12 de Octubre Research Institute (imas12), Madrid, Spain
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Mitochondrial Disorders Laboratory, Clinical Biochemistry Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Dino Gobelli
- Department of Cell Biology, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, Valladolid, Spain
- Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), Valladolid, Spain
| | - Rocío Garrido-Moraga
- Hospital 12 de Octubre Research Institute (imas12), Madrid, Spain
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Mitochondrial Disorders Laboratory, Clinical Biochemistry Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - María J. Esteban-Amo
- Department of Cell Biology, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, Valladolid, Spain
- Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), Valladolid, Spain
| | - José R. López-López
- Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), Valladolid, Spain
- Department of Department of Biochemistry and Molecular Biology and Physiology, Faculty of Medicine, University of Valladolid, Valladolid, Spain
| | - Antonio Orduña
- Division of Microbiology, Hospital Clínico of Valladolid, Valladolid, Spain
- Department of Microbiology, University of Valladolid, Valladolid, Spain
| | - Miguel A. de la Fuente
- Department of Cell Biology, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, Valladolid, Spain
- Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), Valladolid, Spain
| | - Miguel A. Martín
- Hospital 12 de Octubre Research Institute (imas12), Madrid, Spain
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Mitochondrial Disorders Laboratory, Clinical Biochemistry Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - María Simarro
- Department of Cell Biology, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, Valladolid, Spain
- Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), Valladolid, Spain
| |
Collapse
|
20
|
Gilmore LA, Parry TL, Thomas GA, Khamoui AV. Skeletal muscle omics signatures in cancer cachexia: perspectives and opportunities. J Natl Cancer Inst Monogr 2023; 2023:30-42. [PMID: 37139970 PMCID: PMC10157770 DOI: 10.1093/jncimonographs/lgad006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/13/2023] [Accepted: 02/06/2023] [Indexed: 05/05/2023] Open
Abstract
Cachexia is a life-threatening complication of cancer that occurs in up to 80% of patients with advanced cancer. Cachexia reflects the systemic consequences of cancer and prominently features unintended weight loss and skeletal muscle wasting. Cachexia impairs cancer treatment tolerance, lowers quality of life, and contributes to cancer-related mortality. Effective treatments for cancer cachexia are lacking despite decades of research. High-throughput omics technologies are increasingly implemented in many fields including cancer cachexia to stimulate discovery of disease biology and inform therapy choice. In this paper, we present selected applications of omics technologies as tools to study skeletal muscle alterations in cancer cachexia. We discuss how comprehensive, omics-derived molecular profiles were used to discern muscle loss in cancer cachexia compared with other muscle-wasting conditions, to distinguish cancer cachexia from treatment-related muscle alterations, and to reveal severity-specific mechanisms during the progression of cancer cachexia from early toward severe disease.
Collapse
Affiliation(s)
- L Anne Gilmore
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Traci L Parry
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, NC, USA
| | - Gwendolyn A Thomas
- Department of Kinesiology, Pennsylvania State University, University Park, PA, USA
| | - Andy V Khamoui
- Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL, USA
- Institute for Human Health and Disease Intervention, Florida Atlantic University, Jupiter, FL, USA
| |
Collapse
|
21
|
Wall SW, Sanchez L, Tuttle KS, Pearson SJ, Soma S, Wyatt GL, Carter HN, Jenschke RM, Tan L, Martinez SA, Lorenzi PL, Gohil VM, Rijnkels M, Porter WW. Noncanonical role of singleminded-2s in mitochondrial respiratory chain formation in breast cancer. Exp Mol Med 2023; 55:1046-1063. [PMID: 37121978 PMCID: PMC10238511 DOI: 10.1038/s12276-023-00996-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 05/02/2023] Open
Abstract
Dysregulation of cellular metabolism is a hallmark of breast cancer progression and is associated with metastasis and therapeutic resistance. Here, we show that the breast tumor suppressor gene SIM2 promotes mitochondrial oxidative phosphorylation (OXPHOS) using breast cancer cell line models. Mechanistically, we found that SIM2s functions not as a transcription factor but localizes to mitochondria and directly interacts with the mitochondrial respiratory chain (MRC) to facilitate functional supercomplex (SC) formation. Loss of SIM2s expression disrupts SC formation through destabilization of MRC Complex III, leading to inhibition of electron transport, although Complex I (CI) activity is retained. A metabolomic analysis showed that knockout of SIM2s leads to a compensatory increase in ATP production through glycolysis and accelerated glutamine-driven TCA cycle production of NADH, creating a favorable environment for high cell proliferation. Our findings indicate that SIM2s is a novel stabilizing factor required for SC assembly, providing insight into the impact of the MRC on metabolic adaptation and breast cancer progression.
Collapse
Affiliation(s)
- Steven W Wall
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Lilia Sanchez
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | | | - Scott J Pearson
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Shivatheja Soma
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843, USA
| | - Garhett L Wyatt
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Hannah N Carter
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Ramsey M Jenschke
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Sara A Martinez
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Philip L Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Vishal M Gohil
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843, USA
| | - Monique Rijnkels
- Department of Veterinary Integrative Biosciences, School of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA
| | - Weston W Porter
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
22
|
Shen J, Zha Q, Yang QH, Zhou YQ, Liang X, Chen YJ, Qi GX, Zhang XJ, Yao WB, Gao XD, Chen S. Inhibiting von Hippel‒Lindau protein-mediated Dishevelled ubiquitination protects against experimental parkinsonism. Acta Pharmacol Sin 2023; 44:940-953. [PMID: 36357669 PMCID: PMC10104824 DOI: 10.1038/s41401-022-01014-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 10/16/2022] [Indexed: 11/11/2022] Open
Abstract
Dopaminergic neuron degeneration is a hallmark of Parkinson's disease (PD). We previously reported that the inactivation of von Hippel‒Lindau (VHL) alleviated dopaminergic neuron degeneration in a C. elegans model. In this study, we investigated the specific effects of VHL loss and the underlying mechanisms in mammalian PD models. For in vivo genetic inhibition of VHL, AAV-Vhl-shRNA was injected into mouse lateral ventricles. Thirty days later, the mice received MPTP for 5 days to induce PD. Behavioral experiments were conducted on D1, D3, D7, D14 and D21 after the last injection, and the mice were sacrificed on D22. We showed that knockdown of VHL in mice significantly alleviated PD-like syndromes detected in behavioral and biochemical assays. Inhibiting VHL exerted similar protective effects in MPP+-treated differentiated SH-SY5Y cells and the MPP+-induced C. elegans PD model. We further demonstrated that VHL loss-induced protection against experimental parkinsonism was independent of hypoxia-inducible factor and identified the Dishevelled-2 (DVL-2)/β-catenin axis as the target of VHL, which was evolutionarily conserved in both C. elegans and mammals. Inhibiting the function of VHL promoted the stability of β-catenin by reducing the ubiquitination and degradation of DVL-2. Thus, in vivo overexpression of DVL-2, mimicking VHL inactivation, protected against PD. We designed a competing peptide, Tat-DDF-2, to inhibit the interaction between VHL and DVL-2, which exhibited pharmacological potential for protection against PD in vitro and in vivo. We propose the therapeutic potential of targeting the interaction between VHL and DVL-2, which may represent a strategy to alleviate neurodegeneration associated with PD.
Collapse
Affiliation(s)
- Jie Shen
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Qian Zha
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Qian-Hua Yang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Yue-Qian Zhou
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiao Liang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Ying-Jie Chen
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Gui-Xia Qi
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiao-Jin Zhang
- Department of Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Wen-Bing Yao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
| | - Xiang-Dong Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
| | - Song Chen
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
23
|
Dar GM, Ahmad E, Ali A, Mahajan B, Ashraf GM, Saluja SS. Genetic aberration analysis of mitochondrial respiratory complex I implications in the development of neurological disorders and their clinical significance. Ageing Res Rev 2023; 87:101906. [PMID: 36905963 DOI: 10.1016/j.arr.2023.101906] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 03/02/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023]
Abstract
Growing neurological diseases pose difficult challenges for modern medicine to diagnose and manage them effectively. Many neurological disorders mainly occur due to genetic alteration in genes encoding mitochondrial proteins. Moreover, mitochondrial genes exhibit a higher rate of mutation due to the generation of Reactive oxygen species (ROS) during oxidative phosphorylation operating in their vicinity. Among the different complexes of Electron transport chain (ETC), NADH: Ubiquinone oxidoreductase (Mitochondrial complex I) is the most important. This multimeric enzyme, composed of 44 subunits, is encoded by both nuclear and mitochondrial genes. It often exhibits mutations resulting in development of various neurological diseases. The most prominent diseases include leigh syndrome (LS), leber hereditary optic neuropathy (LHON), mitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes (MELAS), myoclonic epilepsy associated with ragged-red fibers (MERRF), idiopathic Parkinson's disease (PD) and, Alzheimer's disease (AD). Preliminary data suggest that mitochondrial complex I subunit genes mutated are frequently of nuclear origin; however, most of the mtDNA gene encoding subunits are also primarily involved. In this review, we have discussed the genetic origins of neurological disorders involving mitochondrial complex I and signified recent approaches to unravel the diagnostic and therapeutic potentials and their management.
Collapse
Affiliation(s)
- Ghulam Mehdi Dar
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi 110002, India
| | - Ejaj Ahmad
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi 110002, India
| | - Asgar Ali
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi 110002, India
| | - Bhawna Mahajan
- Department of Biochemistry, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi 110002, India
| | - Ghulam Md Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, and Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Sundeep Singh Saluja
- Central Molecular Laboratory, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi 110002, India; Department of GI Surgery, Govind Ballabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi 110002, India.
| |
Collapse
|
24
|
Pearson-Smith JN, Fulton R, Huynh CQ, Figueroa AG, Huynh GB, Liang LP, Gano LB, Michel CR, Reisdorph N, Reisdorph R, Fritz KS, Verdin E, Patel M. Neuronal SIRT3 Deletion Predisposes to Female-Specific Alterations in Cellular Metabolism, Memory, and Network Excitability. J Neurosci 2023; 43:1845-1857. [PMID: 36759193 PMCID: PMC10010453 DOI: 10.1523/jneurosci.1259-22.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 01/14/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
Mitochondrial dysfunction is an early event in the pathogenesis of neurologic disorders and aging. Sirtuin 3 (SIRT3) regulates mitochondrial function in response to the cellular environment through the reversible deacetylation of proteins involved in metabolism and reactive oxygen species detoxification. As the primary mitochondrial deacetylase, germline, or peripheral tissue-specific deletion of SIRT3 produces mitochondrial hyperacetylation and the accelerated development of age-related diseases. Given the unique metabolic demands of neurons, the role of SIRT3 in the brain is only beginning to emerge. Using mass spectrometry-based acetylomics, high-resolution respirometry, video-EEG, and cognition testing, we report targeted deletion of SIRT3 from select neurons in the cortex and hippocampus produces altered neuronal excitability and metabolic dysfunction in female mice. Targeted deletion of SIRT3 from neuronal helix-loop-helix 1 (NEX)-expressing neurons resulted in mitochondrial hyperacetylation, female-specific superoxide dismutase-2 (SOD2) modification, increased steady-state superoxide levels, metabolic reprogramming, altered neuronal excitability, and working spatial memory deficits. Inducible neuronal deletion of SIRT3 likewise produced female-specific deficits in spatial working memory. Together, the data demonstrate that deletion of SIRT3 from forebrain neurons selectively predisposes female mice to deficits in mitochondrial and cognitive function.SIGNIFICANCE STATEMENT Mitochondrial SIRT3 is an enzyme shown to regulate energy metabolism and antioxidant function, by direct deacetylation of proteins. In this study, we show that neuronal SIRT3 deficiency renders female mice selectively vulnerable to impairment in redox and metabolic function, spatial memory, and neuronal excitability. The observed sex-specific effects on cognition and neuronal excitability in female SIRT3-deficient mice suggest that mitochondrial dysfunction may be one factor underlying comorbid neuronal diseases, such as Alzheimer's disease and epilepsy. Furthermore, the data suggest that SIRT3 dysfunction may predispose females to age-related metabolic and cognitive impairment.
Collapse
Affiliation(s)
- Jennifer N Pearson-Smith
- School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Ruth Fulton
- School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Christopher Q Huynh
- School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Anna G Figueroa
- School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Gia B Huynh
- School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Li-Ping Liang
- School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Lindsey B Gano
- School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Cole R Michel
- School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Nichole Reisdorph
- School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Richard Reisdorph
- School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Kristofer S Fritz
- School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Eric Verdin
- Buck Institute for Aging, Novato, California 94945
| | - Manisha Patel
- School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
25
|
Marx C, Marx-Blümel L, Sonnemann J, Wang ZQ. Assessment of Mitochondrial Dysfunctions After Sirtuin Inhibition. Methods Mol Biol 2023; 2589:269-291. [PMID: 36255631 DOI: 10.1007/978-1-0716-2788-4_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Posttranslational modifications are important for protein functions and cellular signaling pathways. The acetylation of lysine residues is catalyzed by histone acetyltransferases (HATs) and removed by histone deacetylases (HDACs), with the latter being grouped into four phylogenetic classes. The class III of the HDAC family, the sirtuins (SIRTs), contributes to gene expression, genomic stability, cell metabolism, and tumorigenesis. Thus, several specific SIRT inhibitors (SIRTi) have been developed to target cancer cell proliferation. Here we provide an overview of methods to study SIRT-dependent cell metabolism and mitochondrial functionality. The chapter describes metabolic flux analysis using Seahorse analyzers, methods for normalization of Seahorse data, flow cytometry and fluorescence microscopy to determine the mitochondrial membrane potential, mitochondrial content per cell and mitochondrial network structures, and Western blot analysis to measure mitochondrial proteins.
Collapse
Affiliation(s)
- Christian Marx
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.
| | - Lisa Marx-Blümel
- Department of Pediatric Hematology and Oncology, Children's Clinic, Jena University Hospital, Jena, Germany
- Research Center Lobeda, Jena University Hospital, Jena, Germany
| | - Jürgen Sonnemann
- Department of Pediatric Hematology and Oncology, Children's Clinic, Jena University Hospital, Jena, Germany
- Research Center Lobeda, Jena University Hospital, Jena, Germany
| | - Zhao-Qi Wang
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
- Faculty of Biological Sciences, Friedrich-Schiller-University of Jena, Jena, Germany
| |
Collapse
|
26
|
Schiller J, Laube E, Wittig I, Kühlbrandt W, Vonck J, Zickermann V. Insights into complex I assembly: Function of NDUFAF1 and a link with cardiolipin remodeling. SCIENCE ADVANCES 2022; 8:eadd3855. [PMID: 36383672 PMCID: PMC9668296 DOI: 10.1126/sciadv.add3855] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/14/2022] [Indexed: 06/02/2023]
Abstract
Respiratory complex I is a ~1-MDa proton pump in mitochondria. Its structure has been revealed in great detail, but the structural basis of its assembly, in humans involving at least 15 assembly factors, is essentially unknown. We determined cryo-electron microscopy structures of assembly intermediates associated with assembly factor NDUFAF1 in a yeast model system. Subunits ND2 and NDUFC2 together with assembly factors NDUFAF1 and CIA84 form the nucleation point of the NDUFAF1-dependent assembly pathway. Unexpectedly, the cardiolipin remodeling enzyme tafazzin is an integral component of this core complex. In a later intermediate, all 12 subunits of the proximal proton pump module have assembled. NDUFAF1 locks the central ND3 subunit in an assembly-competent conformation, and major rearrangements of central subunits are required for complex I maturation.
Collapse
Affiliation(s)
- Jonathan Schiller
- Institute of Biochemistry II, University Hospital, Goethe University, 60590 Frankfurt am Main, Germany
- Center for Biomolecular Magnetic Resonance, Institute for Biophysical Chemistry, Goethe University, 60438 Frankfurt am Main, Germany
| | - Eike Laube
- Department of Structural Biology, Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Ilka Wittig
- Functional Proteomics, Institute for Cardiovascular Physiology, Goethe University, 60590 Frankfurt am Main, Germany
| | - Werner Kühlbrandt
- Department of Structural Biology, Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Janet Vonck
- Department of Structural Biology, Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Volker Zickermann
- Institute of Biochemistry II, University Hospital, Goethe University, 60590 Frankfurt am Main, Germany
- Center for Biomolecular Magnetic Resonance, Institute for Biophysical Chemistry, Goethe University, 60438 Frankfurt am Main, Germany
| |
Collapse
|
27
|
Sordaria macrospora Sterile Mutant pro34 Is Impaired in Respiratory Complex I Assembly. J Fungi (Basel) 2022; 8:jof8101015. [PMID: 36294581 PMCID: PMC9605262 DOI: 10.3390/jof8101015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/21/2022] Open
Abstract
The formation of fruiting bodies is a highly regulated process that requires the coordinated formation of different cell types. By analyzing developmental mutants, many developmental factors have already been identified. Yet, a complete understanding of fruiting body formation is still lacking. In this study, we analyzed developmental mutant pro34 of the filamentous ascomycete Sordaria macrospora. Genome sequencing revealed a deletion in the pro34 gene encoding a putative mitochondrial complex I assembly factor homologous to Neurospora crassa CIA84. We show that PRO34 is required for fast vegetative growth, fruiting body and ascospore formation. The pro34 transcript undergoes adenosine to inosine editing, a process correlated with sexual development in fruiting body-forming ascomycetes. Fluorescence microscopy and western blot analysis showed that PRO34 is a mitochondrial protein, and blue-native PAGE revealed that the pro34 mutant lacks mitochondrial complex I. Inhibitor experiments revealed that pro34 respires via complexes III and IV, but also shows induction of alternative oxidase, a shunt pathway to bypass complexes III and IV. We discuss the hypothesis that alternative oxidase is induced to prevent retrograde electron transport to complex I intermediates, thereby protecting from oxidative stress.
Collapse
|
28
|
Redox Active α-Lipoic Acid Differentially Improves Mitochondrial Dysfunction in a Cellular Model of Alzheimer and Its Control Cells. Int J Mol Sci 2022; 23:ijms23169186. [PMID: 36012451 PMCID: PMC9409376 DOI: 10.3390/ijms23169186] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 11/22/2022] Open
Abstract
Introduction: Alpha lipoic acid (ALA) is a sulphur-containing organic compound, derived from octanoic acid, and an important cofactor for mitochondrial respiratory enzymes. It has strong antioxidant properties that improve mitochondrial function. We investigated if ALA improves mitochondrial dysfunction in a cellular model of Alzheimer’s disease (AD). Methods: SH-SY5Y-APP695 cells were used as a model for an early stage of AD. Vector-transfected SH-SY5Y-MOCK cells served as controls. Using these cells, we investigated mitochondrial respiration (OXPHOS), mitochondrial membrane potential (MMP), adenosine triphosphate (ATP) production, and citrate synthase activity (CS) in cells treated with ALA. Cells were treated for 24 h with different concentrations of ALA and with or without the complex I inhibitor rotenone. Results: Incubation with ALA showed a significant increase in ATP levels in both SH-SY5Y-APP695 and SH-SY5Y-MOCK cells. MMP levels were elevated in SH-SY5Y-MOCK cells, treatment with rotenone showed a reduction in MMP, which could be partly alleviated after incubation with ALA in SH-SY5Y-MOCK cells. ALA treatment showed significant differences in respiration chain complex activities in SH-SY5Y-MOCK cells. Citrate synthase activity was unaffected. ROS levels were significantly lower in both cell lines treated with ALA. Conclusions: ALA increased the activity of the different complexes of the respiratory chain, and consequently enhanced the MMP, leading to increased ATP levels indicating improved mitochondrial function. ALA only marginally protects from additional rotenone-induced mitochondrial stress.
Collapse
|
29
|
Huang ML, Yen PL, Chang CH, Liao VHC. Chronic di(2-ethylhexyl) phthalate exposure leads to dopaminergic neuron degeneration through mitochondrial dysfunction in C. elegans. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 307:119574. [PMID: 35671892 DOI: 10.1016/j.envpol.2022.119574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/23/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
The plasticizer di(2-ethylhexyl) phthalate (DEHP) is frequently detected in the environment due to the abundance of its use. These levels might be hazardous to human health and ecosystems. Phthalates have been associated with neurological disorders, yet whether chronic DEHP exposure plays a role in Parkinson's disease (PD) or its underlying mechanisms is unknown. We investigated the effects of chronic DEHP exposure less than an environmentally-relevant dose on PD hallmarks, using Caenorhabditis elegans as a model. We show that developmental stage and exposure timing influence DEHP-induced dopaminergic neuron degeneration. In addition, in response to chronic DEHP exposure at 5 mg/L, mitochondrial fragmentation became significantly elevated, reactive oxygen species (ROS) levels increased, and ATP levels decreased, suggesting that mitochondrial dysfunction occurs. Furthermore, the data show that mitochondrial complex I (nuo-1 and gas-1) and complex II (mev-1) are involved in DEHP-induced dopaminergic neuron toxicity. These results suggest that chronic exposure to DEHP at levels less than an environmentally-relevant dose causes dopaminergic neuron degeneration through mitochondrial dysfunction involving mitochondrial complex I and II. Considering the high level of genetic conservation between C. elegans and mammals, chronic DEHP exposure might elevate the risk of developing PD in humans.
Collapse
Affiliation(s)
- Mei-Lun Huang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Pei-Ling Yen
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Chun-Han Chang
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 106, Taiwan
| | - Vivian Hsiu-Chuan Liao
- Department of Bioenvironmental Systems Engineering, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
30
|
Marx C, Sonnemann J, Maddocks ODK, Marx-Blümel L, Beyer M, Hoelzer D, Thierbach R, Maletzki C, Linnebacher M, Heinzel T, Krämer OH. Global metabolic alterations in colorectal cancer cells during irinotecan-induced DNA replication stress. Cancer Metab 2022; 10:10. [PMID: 35787728 PMCID: PMC9251592 DOI: 10.1186/s40170-022-00286-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/09/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Metabolic adaptations can allow cancer cells to survive DNA-damaging chemotherapy. This unmet clinical challenge is a potential vulnerability of cancer. Accordingly, there is an intense search for mechanisms that modulate cell metabolism during anti-tumor therapy. We set out to define how colorectal cancer CRC cells alter their metabolism upon DNA replication stress and whether this provides opportunities to eliminate such cells more efficiently. METHODS We incubated p53-positive and p53-negative permanent CRC cells and short-term cultured primary CRC cells with the topoisomerase-1 inhibitor irinotecan and other drugs that cause DNA replication stress and consequently DNA damage. We analyzed pro-apoptotic mitochondrial membrane depolarization and cell death with flow cytometry. We evaluated cellular metabolism with immunoblotting of electron transport chain (ETC) complex subunits, analysis of mitochondrial mRNA expression by qPCR, MTT assay, measurements of oxygen consumption and reactive oxygen species (ROS), and metabolic flux analysis with the Seahorse platform. Global metabolic alterations were assessed using targeted mass spectrometric analysis of extra- and intracellular metabolites. RESULTS Chemotherapeutics that cause DNA replication stress induce metabolic changes in p53-positive and p53-negative CRC cells. Irinotecan enhances glycolysis, oxygen consumption, mitochondrial ETC activation, and ROS production in CRC cells. This is connected to increased levels of electron transport chain complexes involving mitochondrial translation. Mass spectrometric analysis reveals global metabolic adaptations of CRC cells to irinotecan, including the glycolysis, tricarboxylic acid cycle, and pentose phosphate pathways. P53-proficient CRC cells, however, have a more active metabolism upon DNA replication stress than their p53-deficient counterparts. This metabolic switch is a vulnerability of p53-positive cells to irinotecan-induced apoptosis under glucose-restricted conditions. CONCLUSION Drugs that cause DNA replication stress increase the metabolism of CRC cells. Glucose restriction might improve the effectiveness of classical chemotherapy against p53-positive CRC cells. The topoisomerase-1 inhibitor irinotecan and other chemotherapeutics that cause DNA damage induce metabolic adaptations in colorectal cancer (CRC) cells irrespective of their p53 status. Irinotecan enhances the glycolysis and oxygen consumption in CRC cells to deliver energy and biomolecules necessary for DNA repair and their survival. Compared to p53-deficient cells, p53-proficient CRC cells have a more active metabolism and use their intracellular metabolites more extensively. This metabolic switch creates a vulnerability to chemotherapy under glucose-restricted conditions for p53-positive cells.
Collapse
Affiliation(s)
- Christian Marx
- Department of Toxicology, University Medical Center, Johannes Gutenberg University Mainz, Building 905, Mainz, Germany.
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Institute for Biochemistry and Biophysics, Friedrich Schiller University of Jena, Jena, Germany.
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.
- Current Address: Center for Pandemic Vaccines and Therapeutics (ZEPAI), Paul Ehrlich Institute, Langen, Germany.
| | - Jürgen Sonnemann
- Department of Paediatric Haematology and Oncology, Jena University Hospital, Children's Clinic, Jena, Germany
- Research Center Lobeda, Jena University Hospital, Jena, Germany
| | - Oliver D K Maddocks
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Lisa Marx-Blümel
- Department of Paediatric Haematology and Oncology, Jena University Hospital, Children's Clinic, Jena, Germany
- Research Center Lobeda, Jena University Hospital, Jena, Germany
| | - Mandy Beyer
- Department of Toxicology, University Medical Center, Johannes Gutenberg University Mainz, Building 905, Mainz, Germany
| | - Doerte Hoelzer
- Department of Human Nutrition, Institute of Nutrition, Friedrich Schiller University of Jena, Jena, Germany
- Current address: Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - René Thierbach
- Department of Human Nutrition, Institute of Nutrition, Friedrich Schiller University of Jena, Jena, Germany
| | - Claudia Maletzki
- Molecular Oncology and Immunotherapy, Thoracic, Vascular and Transplantation Surgery, Clinic of General, University of Rostock, VisceralRostock, Germany
- Current address: Department of Medicine, Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock, Germany
| | - Michael Linnebacher
- Molecular Oncology and Immunotherapy, Thoracic, Vascular and Transplantation Surgery, Clinic of General, University of Rostock, VisceralRostock, Germany
| | - Thorsten Heinzel
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Institute for Biochemistry and Biophysics, Friedrich Schiller University of Jena, Jena, Germany
| | - Oliver H Krämer
- Department of Toxicology, University Medical Center, Johannes Gutenberg University Mainz, Building 905, Mainz, Germany.
- Department of Biochemistry, Center for Molecular Biomedicine (CMB), Institute for Biochemistry and Biophysics, Friedrich Schiller University of Jena, Jena, Germany.
| |
Collapse
|
31
|
Mechanistic Insights of Mitochondrial Dysfunction in Amyotrophic Lateral Sclerosis: An Update on a Lasting Relationship. Metabolites 2022; 12:metabo12030233. [PMID: 35323676 PMCID: PMC8951432 DOI: 10.3390/metabo12030233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive loss of the upper and lower motor neurons. Despite the increasing effort in understanding the etiopathology of ALS, it still remains an obscure disease, and no therapies are currently available to halt its progression. Following the discovery of the first gene associated with familial forms of ALS, Cu–Zn superoxide dismutase, it appeared evident that mitochondria were key elements in the onset of the pathology. However, as more and more ALS-related genes were discovered, the attention shifted from mitochondria impairment to other biological functions such as protein aggregation and RNA metabolism. In recent years, mitochondria have again earned central, mechanistic roles in the pathology, due to accumulating evidence of their derangement in ALS animal models and patients, often resulting in the dysregulation of the energetic metabolism. In this review, we first provide an update of the last lustrum on the molecular mechanisms by which the most well-known ALS-related proteins affect mitochondrial functions and cellular bioenergetics. Next, we focus on evidence gathered from human specimens and advance the concept of a cellular-specific mitochondrial “metabolic threshold”, which may appear pivotal in ALS pathogenesis.
Collapse
|
32
|
Ntagwabira F, Trujillo M, McElroy T, Brown T, Simmons P, Sykes D, Allen AR. Piperlongumine as a Neuro-Protectant in Chemotherapy Induced Cognitive Impairment. Int J Mol Sci 2022; 23:2008. [PMID: 35216124 PMCID: PMC8880369 DOI: 10.3390/ijms23042008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 11/16/2022] Open
Abstract
Advances in the early diagnosis and treatment have led to increases in breast cancer survivorship. Survivors report cognitive impairment symptoms such as loss of concentration and learning and memory deficits which significantly reduce the patient's quality of life. Additional therapies are needed to prevent these side effects and, the precise mechanisms of action responsible are not fully elucidated. However, increasing evidence points toward the use of neuroprotective compounds with antioxidants and anti-inflammatory properties as tools for conserving learning and memory. Here, we examine the ability of piperlongumine (PL), an alkaloid known to have anti-inflammatory and antioxidant effects, to play a neuroprotective role in 16-week-old female C57BL/6J mice treated with a common breast cancer regimen of doxorubicin, cyclophosphamide, and docetaxel (TAC). During social memory testing, TAC-treated mice exhibited impairment, while TAC/PL co-treated mice did not exhibit measurable social memory deficits. Proteomics analysis showed ERK1/2 signaling is involved in TAC and TAC/PL co-treatment. Reduced Nrf2 mRNA expression was also observed. mRNA levels of Gria2 were increased in TAC treated mice and reduced in TAC/PL co-treated mice. In this study, PL protects against social memory impairment when co-administered with TAC via multifactorial mechanisms involving oxidative stress and synaptic plasticity.
Collapse
Affiliation(s)
- Fabio Ntagwabira
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA; (F.N.); (M.T.); (T.M.); (T.B.); (P.S.)
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA
- Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA
| | - Madison Trujillo
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA; (F.N.); (M.T.); (T.M.); (T.B.); (P.S.)
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA
- Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA
| | - Taylor McElroy
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA; (F.N.); (M.T.); (T.M.); (T.B.); (P.S.)
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA
- Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA
| | - Taurean Brown
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA; (F.N.); (M.T.); (T.M.); (T.B.); (P.S.)
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA
- Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA
| | - Pilar Simmons
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA; (F.N.); (M.T.); (T.M.); (T.B.); (P.S.)
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA
| | | | - Antiño R. Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA; (F.N.); (M.T.); (T.M.); (T.B.); (P.S.)
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA
- Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Little Rock, AR 72205, USA
| |
Collapse
|
33
|
Zhang X, Farrell JJ, Tong T, Hu J, Zhu C, Wang L, Mayeux R, Haines JL, Pericak‐Vance MA, Schellenberg GD, Lunetta KL, Farrer LA. Association of mitochondrial variants and haplogroups identified by whole exome sequencing with Alzheimer's disease. Alzheimers Dement 2022; 18:294-306. [PMID: 34152079 PMCID: PMC8764625 DOI: 10.1002/alz.12396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Findings regarding the association between mitochondrial DNA (mtDNA) variants and Alzheimer's disease (AD) are inconsistent. METHODS We developed a pipeline for accurate assembly and variant calling in mitochondrial genomes embedded within whole exome sequences (WES) from 10,831 participants from the Alzheimer's Disease Sequencing Project (ADSP). Association of AD risk was evaluated with each mtDNA variant and variants located in 1158 nuclear genes related to mitochondrial function using the SCORE test. Gene-based tests were performed using SKAT-O. RESULTS Analysis of 4220 mtDNA variants revealed study-wide significant association of AD with a rare MT-ND4L variant (rs28709356 C>T; minor allele frequency = 0.002; P = 7.3 × 10-5 ) as well as with MT-ND4L in a gene-based test (P = 6.71 × 10-5 ). Significant association was also observed with a MT-related nuclear gene, TAMM41, in a gene-based test (P = 2.7 × 10-5 ). The expression of TAMM41 was lower in AD cases than controls (P = .00046) or mild cognitive impairment cases (P = .03). DISCUSSION Significant findings in MT-ND4L and TAMM41 provide evidence for a role of mitochondria in AD.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Department of Medicine (Biomedical Genetics)Boston University School of Medicine72 East Concord StreetBostonMassachusetts02118USA
- Department of BiostatisticsBoston University School of Public Health801 Massachusetts Avenue 3rd FloorBostonMassachusetts02118USA
| | - John J. Farrell
- Department of Medicine (Biomedical Genetics)Boston University School of Medicine72 East Concord StreetBostonMassachusetts02118USA
| | - Tong Tong
- Department of Medicine (Biomedical Genetics)Boston University School of Medicine72 East Concord StreetBostonMassachusetts02118USA
| | - Junming Hu
- Department of Medicine (Biomedical Genetics)Boston University School of Medicine72 East Concord StreetBostonMassachusetts02118USA
| | - Congcong Zhu
- Department of Medicine (Biomedical Genetics)Boston University School of Medicine72 East Concord StreetBostonMassachusetts02118USA
| | | | - Li‐San Wang
- Department of Pathology and Laboratory MedicineUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania19104USA
| | - Richard Mayeux
- Department of NeurologyColumbia UniversityNew YorkNew York10032USA
| | - Jonathan L. Haines
- Department of Population and Quantitative Health Sciences Case Western Reserve UniversityClevelandOhio44106USA
| | | | - Gerard D. Schellenberg
- Department of Pathology and Laboratory MedicineUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania19104USA
| | - Kathryn L. Lunetta
- Department of BiostatisticsBoston University School of Public Health801 Massachusetts Avenue 3rd FloorBostonMassachusetts02118USA
| | - Lindsay A. Farrer
- Department of Medicine (Biomedical Genetics)Boston University School of Medicine72 East Concord StreetBostonMassachusetts02118USA
- Department of BiostatisticsBoston University School of Public Health801 Massachusetts Avenue 3rd FloorBostonMassachusetts02118USA
- Department of NeurologyBoston University School of MedicineBostonMassachusetts02118USA
- Department of OphthalmologyBoston University School of MedicineBostonMassachusetts02118USA
- Department of EpidemiologyBoston University School of Public Health715 Albany StreetBostonMassachusetts02118USA
| |
Collapse
|
34
|
Huttula S, Väyrynen H, Helisalmi S, Kytövuori L, Luukkainen L, Hiltunen M, Remes AM, Krüger J. NDUFA1 p.Gly32Arg variant in early-onset dementia. Neurobiol Aging 2022; 114:113-116. [DOI: 10.1016/j.neurobiolaging.2021.09.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 09/14/2021] [Accepted: 09/25/2021] [Indexed: 01/23/2023]
|
35
|
Wang Q, Lu M, Zhu X, Gu X, Zhang T, Xia C, Yang L, Xu Y, Zhou M. Brain Mitochondrial Dysfunction: A Possible Mechanism Links Early Life Anxiety to Alzheimer’s Disease in Later Life. Aging Dis 2022; 13:1127-1145. [PMID: 35855329 PMCID: PMC9286915 DOI: 10.14336/ad.2022.0221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 02/21/2022] [Indexed: 11/01/2022] Open
Affiliation(s)
- Qixue Wang
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mengna Lu
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xinyu Zhu
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xinyi Gu
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ting Zhang
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenyi Xia
- Department of Physiology, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Li Yang
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Xu
- Department of Physiology, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Mingmei Zhou
- Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Correspondence should be addressed to: Dr. Mingmei Zhou, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China. E-mail:
| |
Collapse
|
36
|
Li JL, Lin TY, Chen PL, Guo TN, Huang SY, Chen CH, Lin CH, Chan CC. Mitochondrial Function and Parkinson's Disease: From the Perspective of the Electron Transport Chain. Front Mol Neurosci 2021; 14:797833. [PMID: 34955747 PMCID: PMC8695848 DOI: 10.3389/fnmol.2021.797833] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/18/2021] [Indexed: 12/21/2022] Open
Abstract
Parkinson’s disease (PD) is known as a mitochondrial disease. Some even regarded it specifically as a disorder of the complex I of the electron transport chain (ETC). The ETC is fundamental for mitochondrial energy production which is essential for neuronal health. In the past two decades, more than 20 PD-associated genes have been identified. Some are directly involved in mitochondrial functions, such as PRKN, PINK1, and DJ-1. While other PD-associate genes, such as LRRK2, SNCA, and GBA1, regulate lysosomal functions, lipid metabolism, or protein aggregation, some have been shown to indirectly affect the electron transport chain. The recent identification of CHCHD2 and UQCRC1 that are critical for functions of complex IV and complex III, respectively, provide direct evidence that PD is more than just a complex I disorder. Like UQCRC1 in preventing cytochrome c from release, functions of ETC proteins beyond oxidative phosphorylation might also contribute to the pathogenesis of PD.
Collapse
Affiliation(s)
- Jeng-Lin Li
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.,Division of Neurology, Department of Internal Medicine, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan County, Taiwan
| | - Tai-Yi Lin
- College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Lin Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Ting-Ni Guo
- Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan
| | - Shu-Yi Huang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Chiang Chan
- Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
37
|
Mitochondrial DNA and Alzheimer's disease: a first case-control study of the Tunisian population. Mol Biol Rep 2021; 49:1687-1700. [PMID: 34854014 DOI: 10.1007/s11033-021-06978-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/17/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disorder in humans and presents a major health problem throughout the world. The etiology of AD is complex, and many factors are implicated, including mitochondria. Mitochondrial alteration has been proposed as a possible cause of AD. Therefore, several studies have focused on finding an association between inherited mitochondrial DNA variants and AD onset. METHODS In this study, we looked, for the first time, for a potential association between mitochondrial haplogroups or polymorphisms and AD in the Tunisian population. We also evaluated the distribution of the major genetic risk factor for AD, the apolipoprotein E epsilon 4 (APOE ε4), in this population. In total, 159 single-nucleotide polymorphisms (SNPs) of mitochondrial DNA haplogroups were genotyped in 254 individuals (58 patients and 196 controls). An additional genotyping of APOE ε4 was performed. RESULTS No significant association between mitochondrial haplogroups and AD was found. However, two individual SNPs, A5656G (p = 0.03821, OR = 10.46) and A13759G (p = 0.03719, OR = 10.78), showed a significant association with AD. APOE 4 was confirmed as a risk factor for AD (p = 0.000014). CONCLUSION Our findings may confirm the absence of a relation between mitochondrial haplogroups and AD and support the possible involvement of some inherited variants in the pathogenicity of AD.
Collapse
|
38
|
Blood-based mitochondrial respiratory chain function in major depression. Transl Psychiatry 2021; 11:593. [PMID: 34789750 PMCID: PMC8599473 DOI: 10.1038/s41398-021-01723-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/26/2021] [Accepted: 11/02/2021] [Indexed: 12/23/2022] Open
Abstract
Mitochondrial dysfunction has been implicated in major depressive disorder (MDD). A measure of mitochondrial respiratory chain (RC) enzymatic activity-the Mitochondrial Health Index (MHI)-has previously been found to correlate with stress and emotional states in caregivers. We here report mitochondrial RC activities, mitochondrial DNA copy number (mtDNAcn), and the composite MHI in unmedicated and somatically healthy subjects with MDD (n = 47) and healthy controls (HC) (n = 11). We also explore, in a subset of the MDD sample (n = 33), whether these markers are associated with response to 8 weeks of SSRI treatment. Mitochondrial RC complexes I, II, IV, citrate synthase (CS), mtDNAcn, and the MHI were assayed in peripheral blood mononuclear cells. Treatment response was defined as >50% decrease on the 25-item Hamilton Depression Rating Scale (HRDS-25). There were no significant differences in MHI or any of the mitochondrial markers between MDD subjects and HCs. Compared to SSRI nonresponders, SSRI responders had significantly higher baseline mitochondrial content markers CS (p = 0.02) and mtDNAcn (p = 0.02), and higher complex I activity (p = 0.01). Complex II activity increased significantly over treatment, irrespective of clinical response (p = 0.03). Complex I activity decreased in responders (n = 9), but increased in nonresponders (n = 18) (group x time interaction, p = 0.02). Absolute treatment-associated change in HDRS-25 scores correlated significantly with change in complex I activity between baseline and week 8 (r = 0.47, p = 0.01). Although mitochondrial markers did not distinguish MDD from controls, they did distinguish SSRI responders from nonresponders. If larger studies validate these mitochondrial differences, they may become useful biomarkers and identify new drug targets.
Collapse
|
39
|
Coenzyme Q at the Hinge of Health and Metabolic Diseases. Antioxidants (Basel) 2021; 10:antiox10111785. [PMID: 34829656 PMCID: PMC8615162 DOI: 10.3390/antiox10111785] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/28/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022] Open
Abstract
Coenzyme Q is a unique lipidic molecule highly conserved in evolution and essential to maintaining aerobic metabolism. It is endogenously synthesized in all cells by a very complex pathway involving a group of nuclear genes that share high homology among species. This pathway is tightly regulated at transcription and translation, but also by environment and energy requirements. Here, we review how coenzyme Q reacts within mitochondria to promote ATP synthesis and also integrates a plethora of metabolic pathways and regulates mitochondrial oxidative stress. Coenzyme Q is also located in all cellular membranes and plasma lipoproteins in which it exerts antioxidant function, and its reaction with different extramitochondrial oxidoreductases contributes to regulate the cellular redox homeostasis and cytosolic oxidative stress, providing a key factor in controlling various apoptosis mechanisms. Coenzyme Q levels can be decreased in humans by defects in the biosynthesis pathway or by mitochondrial or cytosolic dysfunctions, leading to a highly heterogeneous group of mitochondrial diseases included in the coenzyme Q deficiency syndrome. We also review the importance of coenzyme Q levels and its reactions involved in aging and age-associated metabolic disorders, and how the strategy of its supplementation has had benefits for combating these diseases and for physical performance in aging.
Collapse
|
40
|
Xia C, Lou B, Fu Z, Mohsen AW, Shen AL, Vockley J, Kim JJP. Molecular mechanism of interactions between ACAD9 and binding partners in mitochondrial respiratory complex I assembly. iScience 2021; 24:103153. [PMID: 34646991 PMCID: PMC8497999 DOI: 10.1016/j.isci.2021.103153] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/09/2021] [Accepted: 09/16/2021] [Indexed: 01/05/2023] Open
Abstract
The dual function protein ACAD9 catalyzes α,β-dehydrogenation of fatty acyl-CoA thioesters in fatty acid β-oxidation and is an essential chaperone for mitochondrial respiratory complex I (CI) assembly. ACAD9, ECSIT, and NDUFAF1 interact to form the core mitochondrial CI assembly complex. Current studies examine the molecular mechanism of ACAD9/ECSIT/NDUFAF1interactions. ACAD9 binds to the carboxy-terminal half and NDUFAF1 to the amino-terminal half of ECSIT. Binary complexes are unstable and aggregate easily, while the ACAD9/ECSIT/NDUFAF1 ternary complex is soluble and highly stable. Molecular modeling and small-angle X-ray scattering studies identified intra-complex interaction sites and binding sites for other assembly factors. Binding of ECSIT at the ETF binding site in the amino-terminal domain of ACAD9 is consistent with observed loss of FAD and enzymatic activity and demonstrates that the two functions of ACAD9 are mutually exclusive. Mapping of 42 known pathogenic mutations onto the homology-modeled ACAD9 structure provides structural insights into pathomechanisms of CI deficiency.
Collapse
Affiliation(s)
- Chuanwu Xia
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Baoying Lou
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Zhuji Fu
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Al-Walid Mohsen
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Anna L. Shen
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jerry Vockley
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jung-Ja P. Kim
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
41
|
Sanz FJ, Solana-Manrique C, Torres J, Masiá E, Vicent MJ, Paricio N. A High-Throughput Chemical Screen in DJ-1β Mutant Flies Identifies Zaprinast as a Potential Parkinson's Disease Treatment. Neurotherapeutics 2021; 18:2565-2578. [PMID: 34697772 PMCID: PMC8804136 DOI: 10.1007/s13311-021-01134-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Dopamine replacement represents the standard therapy for Parkinson's disease (PD), a common, chronic, and incurable neurological disorder; however, this approach only treats the symptoms of this devastating disease. In the search for novel disease-modifying therapies that target other relevant molecular and cellular mechanisms, Drosophila has emerged as a valuable tool to study neurodegenerative diseases due to the presence of a complex central nervous system, the blood-brain barrier, and a similar neurotransmitter profile to humans. Human PD-related genes also display conservation in flies; DJ-1β is the fly ortholog of DJ-1, a gene for which mutations prompt early-onset recessive PD. Interestingly, flies mutant for DJ-1β exhibit PD-related phenotypes, including motor defects, high oxidative stress (OS) levels and metabolic alterations. To identify novel therapies for PD, we performed an in vivo high-throughput screening assay using DJ-1β mutant flies and compounds from the Prestwick® chemical library. Drugs that improved motor performance in DJ-1ß mutant flies were validated in DJ-1-deficient human neural-like cells, revealing that zaprinast displayed the most significant ability to suppress OS-induced cell death. Zaprinast inhibits phosphodiesterases and activates GPR35, an orphan G-protein-coupled receptor not previously associated with PD. We found that zaprinast exerts its beneficial effect in both fly and human PD models through several disease-modifying mechanisms, including reduced OS levels, attenuated apoptosis, increased mitochondrial viability, and enhanced glycolysis. Therefore, our results support zaprinast as a potential therapeutic for PD in future clinical trials.
Collapse
Affiliation(s)
- Francisco José Sanz
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain
- Instituto Universitario de Biotecnología Y Biomedicina (BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain
| | - Cristina Solana-Manrique
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain
- Instituto Universitario de Biotecnología Y Biomedicina (BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain
| | - Josema Torres
- Departamento de Biología Celular, Biología Funcional Y Antropología Física, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain
| | - Esther Masiá
- Polymer Therapeutics Lab and Screening Platform, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - María J Vicent
- Polymer Therapeutics Lab and Screening Platform, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - Nuria Paricio
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain.
- Instituto Universitario de Biotecnología Y Biomedicina (BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain.
| |
Collapse
|
42
|
Hesperetin Nanocrystals Improve Mitochondrial Function in a Cell Model of Early Alzheimer Disease. Antioxidants (Basel) 2021; 10:antiox10071003. [PMID: 34201544 PMCID: PMC8300699 DOI: 10.3390/antiox10071003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 11/21/2022] Open
Abstract
Mitochondrial dysfunction represents a hallmark of both brain aging and age-related neurodegenerative disorders including Alzheimer disease (AD). AD-related mitochondrial dysfunction is characterized by an impaired electron transport chain (ETC), subsequent decreased adenosine triphoshpate (ATP) levels, and elevated generation of reactive oxygen species (ROS). The bioactive citrus flavanone hesperetin (Hst) is known to modulate inflammatory response, to function as an antioxidant, and to provide neuroprotective properties. The efficacy in improving mitochondrial dysfunction of Hst nanocrystals (HstN) with increased bioavailability has not yet been investigated. Human SH-SY5Y cells harboring neuronal amyloid precursor protein (APP695) acted as a model for the initial phase of AD. MOCK-transfected cells served as controls. The energetic metabolite ATP was determined using a luciferase-catalyzed bioluminescence assay. The activity of mitochondrial respiration chain complexes was assessed by high-resolution respirometry using a Clarke electrode. Expression levels of mitochondrial respiratory chain complex genes were determined using quantitative real-time polymerase chain reaction (qRT-PCR). The levels of amyloid β-protein (Aβ1-40) were measured using homogeneous time-resolved fluorescence (HTRF). ROS levels, peroxidase activity, and cytochrome c activity were determined using a fluorescence assay. Compared to pure Hst dissolved in ethanol (HstP), SH-SY5Y-APP695 cells incubated with HstN resulted in significantly reduced mitochondrial dysfunction: ATP levels and respiratory chain complex activity significantly increased. Gene expression levels of RCC I, IV, and V were significantly upregulated. In comparison, the effects of HstN on SY5Y-MOCK control cells were relatively small. Pure Hst dissolved in ethanol (HstP) had almost no effect on both cell lines. Neither HstN nor HstP led to significant changes in Aβ1-40 levels. HstN and HstP were both shown to lower peroxidase activity significantly. Furthermore, HstN significantly reduced cytochrome c activity, whereas HstP had a significant effect on reducing ROS in SH-SY5Y-APP695 cells. Thus, it seems that the mechanisms involved may not be linked to altered Aβ production. Nanoflavonoids such as HstN have the potential to prevent mitochondria against dysfunction. Compared to its pure form, HstN showed a greater effect in combatting mitochondrial dysfunction. Further studies should evaluate whether HstN protects against age-related mitochondrial dysfunction and thus may contribute to late-onset AD.
Collapse
|
43
|
Le Vasseur M, Friedman J, Jost M, Xu J, Yamada J, Kampmann M, Horlbeck MA, Salemi MR, Phinney BS, Weissman JS, Nunnari J. Genome-wide CRISPRi screening identifies OCIAD1 as a prohibitin client and regulatory determinant of mitochondrial Complex III assembly in human cells. eLife 2021; 10:67624. [PMID: 34034859 PMCID: PMC8154037 DOI: 10.7554/elife.67624] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/10/2021] [Indexed: 01/01/2023] Open
Abstract
Dysfunction of the mitochondrial electron transport chain (mETC) is a major cause of human mitochondrial diseases. To identify determinants of mETC function, we screened a genome-wide human CRISPRi library under oxidative metabolic conditions with selective inhibition of mitochondrial Complex III and identified ovarian carcinoma immunoreactive antigen (OCIA) domain-containing protein 1 (OCIAD1) as a Complex III assembly factor. We find that OCIAD1 is an inner mitochondrial membrane protein that forms a complex with supramolecular prohibitin assemblies. Our data indicate that OCIAD1 is required for maintenance of normal steady-state levels of Complex III and the proteolytic processing of the catalytic subunit cytochrome c1 (CYC1). In OCIAD1 depleted mitochondria, unprocessed CYC1 is hemylated and incorporated into Complex III. We propose that OCIAD1 acts as an adaptor within prohibitin assemblies to stabilize and/or chaperone CYC1 and to facilitate its proteolytic processing by the IMMP2L protease.
Collapse
Affiliation(s)
- Maxence Le Vasseur
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, Davis, United States
| | - Jonathan Friedman
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, Davis, United States.,Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Marco Jost
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, United States.,Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, United States.,Department of Microbiology and Immunology, University of California at San Francisco, San Francisco, United States
| | - Jiawei Xu
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, Davis, United States
| | - Justin Yamada
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, Davis, United States
| | - Martin Kampmann
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, United States.,Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, United States.,Institute for Neurodegenerative Diseases and Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, United States.,Chan-Zuckerberg Biohub, San Francisco, United States
| | - Max A Horlbeck
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, United States.,Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, United States
| | - Michelle R Salemi
- Proteomics Core Facility, University of California, Davis, Davis, United States
| | - Brett S Phinney
- Proteomics Core Facility, University of California, Davis, Davis, United States
| | - Jonathan S Weissman
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, United States.,Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, United States.,Whitehead Institute, Cambridge, United States.,Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Jodi Nunnari
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, Davis, United States
| |
Collapse
|
44
|
Subrahmanian N, LaVoie MJ. Is there a special relationship between complex I activity and nigral neuronal loss in Parkinson's disease? A critical reappraisal. Brain Res 2021; 1767:147434. [PMID: 33745923 PMCID: PMC9520341 DOI: 10.1016/j.brainres.2021.147434] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/25/2021] [Accepted: 03/12/2021] [Indexed: 12/21/2022]
Abstract
Parkinson’s disease (PD) is a progressive neurodegenerative disease manifesting both motor and non-motor symptoms. The motor features are generally ascribed to the selective loss of dopamine neurons within the substantia nigra pars compacta. While the precise etiology of PD remains elusive, multiple genetic and environmental elements have emerged as contributing factors. The discovery of MPTP-induced parkinsonism directed intense inquiry towards mitochondrial pathways, with a specific focus on mitochondrial complex I. Consisting of more than 40 subunits, complex I is the first enzyme of the electron transport chain that is required for mitochondrial ATP production. In this review, we present a critical analysis of studies assessing the prevalence and specificity of mitochondrial complex I deficiency in PD. In addition, we take the novel view of incorporating the features of genetically-defined bona fide complex I disorders and the prevalence of nigral involvement in such cases. Through this innovative bi-directional view, we consider both complex I changes in a disease of the substantia nigra and nigral changes in diseases of complex I. We assess the strength of association between nigral cell loss and complex I deficits, as well as the oft under-appreciated heterogeneity of complex I deficiency disorders and the variability of the PD data.
Collapse
Affiliation(s)
- Nitya Subrahmanian
- Department of Neurology, University of Florida, Gainesville, FL 32610, USA
| | - Matthew J LaVoie
- Department of Neurology, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
45
|
Giachin G, Jessop M, Bouverot R, Acajjaoui S, Saïdi M, Chretien A, Bacia‐Verloop M, Signor L, Mas PJ, Favier A, Borel Meneroud E, Hons M, Hart DJ, Kandiah E, Boeri Erba E, Buisson A, Leonard G, Gutsche I, Soler‐Lopez M. Assembly of The Mitochondrial Complex I Assembly Complex Suggests a Regulatory Role for Deflavination. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202011548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Gabriele Giachin
- Structural Biology Group European Synchrotron Radiation Facility (ESRF) 71 avenue des Martyrs 38043 Grenoble France
| | - Matthew Jessop
- Institut de Biologie Structurale (IBS) CNRS, CEA Université Grenoble Alpes 71 avenue des Martyrs 38044 Grenoble France
| | - Romain Bouverot
- Structural Biology Group European Synchrotron Radiation Facility (ESRF) 71 avenue des Martyrs 38043 Grenoble France
| | - Samira Acajjaoui
- Structural Biology Group European Synchrotron Radiation Facility (ESRF) 71 avenue des Martyrs 38043 Grenoble France
| | - Melissa Saïdi
- Structural Biology Group European Synchrotron Radiation Facility (ESRF) 71 avenue des Martyrs 38043 Grenoble France
| | - Anaïs Chretien
- Structural Biology Group European Synchrotron Radiation Facility (ESRF) 71 avenue des Martyrs 38043 Grenoble France
| | - Maria Bacia‐Verloop
- Institut de Biologie Structurale (IBS) CNRS, CEA Université Grenoble Alpes 71 avenue des Martyrs 38044 Grenoble France
| | - Luca Signor
- Institut de Biologie Structurale (IBS) CNRS, CEA Université Grenoble Alpes 71 avenue des Martyrs 38044 Grenoble France
| | - Philippe J. Mas
- Integrated Structural Biology Grenoble (ISBG) CNRS CEA, Université Grenoble Alpes 71 avenue des Martyrs 38042 Grenoble France
| | - Adrien Favier
- Institut de Biologie Structurale (IBS) CNRS, CEA Université Grenoble Alpes 71 avenue des Martyrs 38044 Grenoble France
| | - Eve Borel Meneroud
- Grenoble Institut des Neurosciences (GIN) Centre Inserm U1216 Equipe Neuropathologies et Dysfonctions Synaptiques Université Grenoble Alpes 31 Chemin Fortuné Ferrini 38700 La Tronche France
| | - Michael Hons
- European Molecular Biology Laboratory (EMBL) Grenoble Outstation 71 avenue des Martyrs 38042 Grenoble France
| | - Darren J. Hart
- Institut de Biologie Structurale (IBS) CNRS, CEA Université Grenoble Alpes 71 avenue des Martyrs 38044 Grenoble France
| | - Eaazhisai Kandiah
- Structural Biology Group European Synchrotron Radiation Facility (ESRF) 71 avenue des Martyrs 38043 Grenoble France
| | - Elisabetta Boeri Erba
- Institut de Biologie Structurale (IBS) CNRS, CEA Université Grenoble Alpes 71 avenue des Martyrs 38044 Grenoble France
| | - Alain Buisson
- Grenoble Institut des Neurosciences (GIN) Centre Inserm U1216 Equipe Neuropathologies et Dysfonctions Synaptiques Université Grenoble Alpes 31 Chemin Fortuné Ferrini 38700 La Tronche France
| | - Gordon Leonard
- Structural Biology Group European Synchrotron Radiation Facility (ESRF) 71 avenue des Martyrs 38043 Grenoble France
| | - Irina Gutsche
- Institut de Biologie Structurale (IBS) CNRS, CEA Université Grenoble Alpes 71 avenue des Martyrs 38044 Grenoble France
| | - Montserrat Soler‐Lopez
- Structural Biology Group European Synchrotron Radiation Facility (ESRF) 71 avenue des Martyrs 38043 Grenoble France
| |
Collapse
|
46
|
Albuck AL, Sakamuri SSVP, Sperling JA, Evans WR, Kolli L, Sure VN, Mostany R, Katakam PVG. Peroxynitrite decomposition catalyst enhances respiratory function in isolated brain mitochondria. Am J Physiol Heart Circ Physiol 2021; 320:H630-H641. [PMID: 33164581 PMCID: PMC8082788 DOI: 10.1152/ajpheart.00389.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 10/07/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023]
Abstract
Peroxynitrite (PN), generated from the reaction of nitric oxide (NO) and superoxide, is implicated in the pathogenesis of ischemic and neurodegenerative brain injuries. Mitochondria produce NO from mitochondrial NO synthases and superoxide by the electron transport chain. Our objective was to detect the generation of PN of mitochondrial origin and characterize its effects on mitochondrial respiratory function. Freshly isolated brain nonsynaptosomal mitochondria from C57Bl/6 (wild type, WT) and endothelial NO synthase knockout (eNOS-KO) mice were treated with exogenous PN (0.1, 1, 5 µmol/L) or a PN donor (SIN-1; 50 µmol/L) or a PN scavenger (FeTMPyP; 2.5 µmol/L). Oxygen consumption rate (OCR) was measured using Agilent Seahorse XFe24 analyzer and mitochondrial respiratory parameters were calculated. Mitochondrial membrane potential, superoxide, and PN were determined from rhodamine 123, dihydroethidium, and DAX-J2 PON green fluorescence measurements, respectively. Mitochondrial protein nitrotyrosination was determined by Western blots. Both exogenous PN and SIN-1 decreased respiratory function in WT isolated brain mitochondria. FeTMPyP enhanced state III and state IVo mitochondrial respiration in both WT and eNOS-KO mitochondria. FeTMPyP also elevated state IIIu respiration in eNOS-KO mitochondria. Unlike PN, neither SIN-1 nor FeTMPyP depolarized the mitochondria. Although mitochondrial protein nitrotyrosination was unaffected by SIN-1 or FeTMPyP, FeTMPyP reduced mitochondrial PN levels. Mitochondrial superoxide levels were increased by FeTMPyP but were unaffected by PN or SIN-1. Thus, we present the evidence of functionally significant PN generation in isolated brain mitochondria. Mitochondrial PN activity was physiologically relevant in WT mice and pathologically significant under conditions with eNOS deficiency.NEW & NOTEWORTHY Mitochondria generate superoxide and nitric oxide that could potentially react with each other to produce PN. We observed eNOS and nNOS immunoreactivity in isolated brain and heart mitochondria with pharmacological inhibition of nNOS found to modulate the mitochondrial respiratory function. This study provides evidence of generation of functionally significant PN in isolated brain mitochondria that affects respiratory function under physiological conditions. Importantly, the mitochondrial PN levels and activity were exaggerated in the eNOS-deficient mice, suggesting its pathological significance.
Collapse
Affiliation(s)
- Aaron L Albuck
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Brain Institute, Tulane University, New Orleans, Louisiana
| | - Siva S V P Sakamuri
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Jared A Sperling
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Wesley R Evans
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Brain Institute, Tulane University, New Orleans, Louisiana
| | - Lahari Kolli
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Venkata N Sure
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Ricardo Mostany
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Brain Institute, Tulane University, New Orleans, Louisiana
| | - Prasad V G Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Brain Institute, Tulane University, New Orleans, Louisiana
- Clinical Neuroscience Research Center, New Orleans, Louisiana
| |
Collapse
|
47
|
Giachin G, Jessop M, Bouverot R, Acajjaoui S, Saïdi M, Chretien A, Bacia-Verloop M, Signor L, Mas PJ, Favier A, Borel Meneroud E, Hons M, Hart DJ, Kandiah E, Boeri Erba E, Buisson A, Leonard G, Gutsche I, Soler-Lopez M. Assembly of The Mitochondrial Complex I Assembly Complex Suggests a Regulatory Role for Deflavination. Angew Chem Int Ed Engl 2021; 60:4689-4697. [PMID: 33320993 PMCID: PMC7986633 DOI: 10.1002/anie.202011548] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Indexed: 01/01/2023]
Abstract
Fatty acid β‐oxidation (FAO) and oxidative phosphorylation (OXPHOS) are mitochondrial redox processes that generate ATP. The biogenesis of the respiratory Complex I, a 1 MDa multiprotein complex that is responsible for initiating OXPHOS, is mediated by assembly factors including the mitochondrial complex I assembly (MCIA) complex. However, the organisation and the role of the MCIA complex are still unclear. Here we show that ECSIT functions as the bridging node of the MCIA core complex. Furthermore, cryo‐electron microscopy together with biochemical and biophysical experiments reveal that the C‐terminal domain of ECSIT directly binds to the vestigial dehydrogenase domain of the FAO enzyme ACAD9 and induces its deflavination, switching ACAD9 from its role in FAO to an MCIA factor. These findings provide the structural basis for the MCIA complex architecture and suggest a unique molecular mechanism for coordinating the regulation of the FAO and OXPHOS pathways to ensure an efficient energy production.
Collapse
Affiliation(s)
- Gabriele Giachin
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 71 avenue des Martyrs, 38043, Grenoble, France
| | - Matthew Jessop
- Institut de Biologie Structurale (IBS), CNRS, CEA, Université Grenoble Alpes, 71 avenue des Martyrs, 38044, Grenoble, France
| | - Romain Bouverot
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 71 avenue des Martyrs, 38043, Grenoble, France
| | - Samira Acajjaoui
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 71 avenue des Martyrs, 38043, Grenoble, France
| | - Melissa Saïdi
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 71 avenue des Martyrs, 38043, Grenoble, France
| | - Anaïs Chretien
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 71 avenue des Martyrs, 38043, Grenoble, France
| | - Maria Bacia-Verloop
- Institut de Biologie Structurale (IBS), CNRS, CEA, Université Grenoble Alpes, 71 avenue des Martyrs, 38044, Grenoble, France
| | - Luca Signor
- Institut de Biologie Structurale (IBS), CNRS, CEA, Université Grenoble Alpes, 71 avenue des Martyrs, 38044, Grenoble, France
| | - Philippe J Mas
- Integrated Structural Biology Grenoble (ISBG) CNRS, CEA, Université Grenoble Alpes, 71 avenue des Martyrs, 38042, Grenoble, France
| | - Adrien Favier
- Institut de Biologie Structurale (IBS), CNRS, CEA, Université Grenoble Alpes, 71 avenue des Martyrs, 38044, Grenoble, France
| | - Eve Borel Meneroud
- Grenoble Institut des Neurosciences (GIN), Centre Inserm U1216, Equipe Neuropathologies et Dysfonctions Synaptiques, Université Grenoble Alpes, 31 Chemin Fortuné Ferrini, 38700, La Tronche, France
| | - Michael Hons
- European Molecular Biology Laboratory (EMBL), Grenoble Outstation, 71 avenue des Martyrs, 38042, Grenoble, France
| | - Darren J Hart
- Institut de Biologie Structurale (IBS), CNRS, CEA, Université Grenoble Alpes, 71 avenue des Martyrs, 38044, Grenoble, France
| | - Eaazhisai Kandiah
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 71 avenue des Martyrs, 38043, Grenoble, France
| | - Elisabetta Boeri Erba
- Institut de Biologie Structurale (IBS), CNRS, CEA, Université Grenoble Alpes, 71 avenue des Martyrs, 38044, Grenoble, France
| | - Alain Buisson
- Grenoble Institut des Neurosciences (GIN), Centre Inserm U1216, Equipe Neuropathologies et Dysfonctions Synaptiques, Université Grenoble Alpes, 31 Chemin Fortuné Ferrini, 38700, La Tronche, France
| | - Gordon Leonard
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 71 avenue des Martyrs, 38043, Grenoble, France
| | - Irina Gutsche
- Institut de Biologie Structurale (IBS), CNRS, CEA, Université Grenoble Alpes, 71 avenue des Martyrs, 38044, Grenoble, France
| | - Montserrat Soler-Lopez
- Structural Biology Group, European Synchrotron Radiation Facility (ESRF), 71 avenue des Martyrs, 38043, Grenoble, France
| |
Collapse
|
48
|
Mitochondrial Structure and Bioenergetics in Normal and Disease Conditions. Int J Mol Sci 2021; 22:ijms22020586. [PMID: 33435522 PMCID: PMC7827222 DOI: 10.3390/ijms22020586] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are ubiquitous intracellular organelles found in almost all eukaryotes and involved in various aspects of cellular life, with a primary role in energy production. The interest in this organelle has grown stronger with the discovery of their link to various pathologies, including cancer, aging and neurodegenerative diseases. Indeed, dysfunctional mitochondria cannot provide the required energy to tissues with a high-energy demand, such as heart, brain and muscles, leading to a large spectrum of clinical phenotypes. Mitochondrial defects are at the origin of a group of clinically heterogeneous pathologies, called mitochondrial diseases, with an incidence of 1 in 5000 live births. Primary mitochondrial diseases are associated with genetic mutations both in nuclear and mitochondrial DNA (mtDNA), affecting genes involved in every aspect of the organelle function. As a consequence, it is difficult to find a common cause for mitochondrial diseases and, subsequently, to offer a precise clinical definition of the pathology. Moreover, the complexity of this condition makes it challenging to identify possible therapies or drug targets.
Collapse
|
49
|
Gueguen N, Lenaers G, Reynier P, Weissig V, Edeas M. Mitochondrial Dysfunction in Mitochondrial Medicine: Current Limitations, Pitfalls, and Tomorrow. Methods Mol Biol 2021; 2276:1-29. [PMID: 34060029 DOI: 10.1007/978-1-0716-1266-8_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Until recently restricted to hereditary mitochondrial diseases, mitochondrial dysfunction is now recognized as a key player and strategic factor in the pathophysiological of many human diseases, ranging from the metabolism, vascular, cardiac, and neurodegenerative diseases to cancer. Because of their participation in a myriad of cellular functions and signaling pathways, precisely identifying the cause of mitochondrial "dysfunctions" can be challenging and requires robust and controlled techniques. Initially limited to the analysis of the respiratory chain functioning, these analytical techniques now enlarge to the analyses of mitochondrial and cellular metabolism, based on metabolomic approaches.Here, we address the methods used to assay mitochondrial dysfunction, with a highlight on the techniques used in diagnosis on tissues and cells derived from patients, the information they provide, and their strength and weakness.Targeting mitochondrial dysfunction by various strategies is a huge challenge, requires robust methods of evaluation, and should be able to take into consideration the mitochondria dynamics and localization. The future of mitochondrial medicine is strongly related to a perfect comprehension of its dysfunction.
Collapse
Affiliation(s)
- Naig Gueguen
- UMR CNRS 6015-INSERM U1083, MitoVasc Institute, University of Angers, Angers, France.,Department of Biochemistry and Genetics, University Hospital of Angers, Angers, France
| | - Guy Lenaers
- UMR CNRS 6015-INSERM U1083, MitoVasc Institute, University of Angers, Angers, France
| | - Pascal Reynier
- UMR CNRS 6015-INSERM U1083, MitoVasc Institute, University of Angers, Angers, France.,Department of Biochemistry and Genetics, University Hospital of Angers, Angers, France
| | - Volkmar Weissig
- Department of Pharmaceutical Sciences and Nanocenter of Excellence, Midwestern University College of Pharmacy at Glendale, Glendale, AZ, USA
| | - Marvin Edeas
- Université de Paris, INSERM U1016, Institut Cochin, CNRS UMR8104, Paris, France. .,Laboratory of Excellence GR-Ex, Paris, France.
| |
Collapse
|
50
|
Snow WM, Cadonic C, Cortes-Perez C, Adlimoghaddam A, Roy Chowdhury SK, Thomson E, Anozie A, Bernstein MJ, Gough K, Fernyhough P, Suh M, Albensi BC. Sex-Specific Effects of Chronic Creatine Supplementation on Hippocampal-Mediated Spatial Cognition in the 3xTg Mouse Model of Alzheimer's Disease. Nutrients 2020; 12:nu12113589. [PMID: 33238473 PMCID: PMC7700653 DOI: 10.3390/nu12113589] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/23/2022] Open
Abstract
The creatine (Cr) energy system has been implicated in Alzheimer’s disease (AD), including reductions in brain phosphoCr and Cr kinase, yet no studies have examined the neurobehavioral effects of Cr supplementation in AD, including the 3xTg mouse model. This studied investigated the effects of Cr supplementation on spatial cognition, plasticity- and disease-related protein levels, and mitochondrial function in the 3xTg hippocampus. Here, 3xTg mice were fed a control or Cr-supplemented (3% Cr (w/w)) diet for 8–9 weeks and tested in the Morris water maze. Mitochondrial oxygen consumption (Seahorse) and protein levels (Western blots) were measured in the hippocampus in subsets of mice. Overall, 3xTg females exhibited impaired memory as compared to males. In females, Cr supplementation decreased escape latency and was associated with increased spatial search strategy use. In males, Cr supplementation decreased the use of spatial search strategies. Pilot data indicated mitochondrial enhancements with Cr supplementation in both sexes. In females, Cr supplementation increased CREB phosphorylation and levels of IκB (NF-κB suppressor), CaMKII, PSD-95, and high-molecular-weight amyloid β (Aβ) species, whereas Aβ trimers were reduced. These data suggest a beneficial preventative effect of Cr supplementation in females and warrant caution against Cr supplementation in males in the AD-like brain.
Collapse
Affiliation(s)
- Wanda M. Snow
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Centre for the Advancement of Teaching and Learning, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Correspondence: or (W.M.S); (B.C.A.); Tel.: +1-204-235-3942 (B.C.A.)
| | - Chris Cadonic
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Claudia Cortes-Perez
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Aida Adlimoghaddam
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Subir K. Roy Chowdhury
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Research Institute in Oncology, CancerCare Manitoba, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Ella Thomson
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Adama Anozie
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
| | - Michael J. Bernstein
- Department of Psychological and Social Sciences, Pennsylvania State University Abington, Abington, PA 19001, USA;
| | - Kathleen Gough
- Department of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada;
| | - Paul Fernyhough
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Miyoung Suh
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Benedict C. Albensi
- Division of Neurodegenerative Disorders, St Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H2A6, Canada; (C.C.); (C.C.-P.); (A.A.); (S.K.R.C.); (E.T.); (A.A.); (P.F.); (M.S.)
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Correspondence: or (W.M.S); (B.C.A.); Tel.: +1-204-235-3942 (B.C.A.)
| |
Collapse
|